Language selection

Search

Patent 3039510 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3039510
(54) English Title: IMMUNOGENIC ARGINASE PEPTIDES
(54) French Title: PEPTIDES D'ARGINASE IMMUNOGENES
Status: Examination Requested
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 38/00 (2006.01)
  • C12N 9/96 (2006.01)
(72) Inventors :
  • ANDERSEN, MADS HALD (Denmark)
(73) Owners :
  • IO BIOTECH APS (Denmark)
(71) Applicants :
  • IO BIOTECH APS (Denmark)
(74) Agent: NORTON ROSE FULBRIGHT CANADA LLP/S.E.N.C.R.L., S.R.L.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2017-10-06
(87) Open to Public Inspection: 2018-04-12
Examination requested: 2022-08-29
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2017/075443
(87) International Publication Number: WO2018/065563
(85) National Entry: 2019-04-04

(30) Application Priority Data:
Application No. Country/Territory Date
16192794.2 European Patent Office (EPO) 2016-10-07

Abstracts

English Abstract

The present invention relates to immunogenic polypeptide fragments of a human Arginase protein. The fragments are in particular useful for the treatment or prevention of cancer.


French Abstract

La présente invention concerne des fragments polypeptidiques immunogènes d'une protéine Arginase humaine. Les fragments sont particulièrement utiles pour le traitement ou la prévention du cancer.

Claims

Note: Claims are shown in the official language in which they were submitted.


50
CLAIMS
1. An isolated, immunogenic polypeptide fragment of a human Arginase
protein of SEQ ID
NO: 1 (Arginase 1) or SEQ ID NO: 60 (Arginase 2), which fragment is up to 55
amino acids in
length and which comprises or consists of a sequence of at least 8, 9, 10, 20,
30, 40 or 50
consecutive amino acids of either of SEQ ID NO: 52 or SEQ ID NO: 58.
2. The polypeptide fragment of claim 1 which comprises or consists of the
amino acid
sequence of any one of SEQ ID NOs: 52, 50, 51, 37, 35, 35, 34, 9, 53, 54, 2 to
33, or 38 to 49.
3. The polypeptide fragment of claim 1 or 2 which comprises at least 8, 9,
10, 15 or all 20
consecutive amino acids of any one of SEQ ID NOs: 37, 36, 34 or 35, optionally
wherein the
cysteine in SEQ ID NO: 34 is replaced by conservative substitution.
4. The polypeptide fragment of any one of the preceding claims, wherein:
a. the C terminal amino acid is replaced with the corresponding amide; and/or
b. the L at the position corresponding to position 190 of SEQ ID NO: 1 is
replaced with
I; and/or
c. the R at the position corresponding to position 205 of SEQ ID NO: 1 is
replaced with
K; and/or
d. at least one additional moiety is attached to the N and/or C terminus to
improve
solubility or manufacturability of the polypeptide, optionally wherein said
additional
moiety is a hydrophilic amino acid such as R or K; and/or
e. said fragment lacks or has reduced arginase activity relative to the
corresponding full
length arginase; and/or
f. said fragment is capable of stimulating T cells which recognize cells
expressing the
corresponding arginase.
5. A composition comprising the polypeptide fragment of any one of claims 1
to 4, a
pharmaceutically acceptable diluent or carrier, and optionally an adjuvant.
6. A composition according to claim 5 wherein the adjuvant is selected from
the group
consisting of bacterial DNA based adjuvants, oil/surfactant based adjuvants,
viral dsRNA based
adjuvants, imidazochinilines, and a Montanide ISA adjuvant.

51
7. A method of treating or preventing a disease or condition in a subject,
the method
comprising administering to the subject the isolated polypeptide of any one of
claims 1 to 4 or
the composition of claim 5 or 6.
8. The method of claim 7 wherein the disease or condition is characterized
at least in part by
inappropriate or excessive immune suppressive function of an Arginase, and/or
said wherein
disease or condition is cancer.
9. The method of claim 8 wherein the disease or condition is cancer and
optionally wherein
the method further comprises the simultaneous or sequential administration of
an additional
cancer therapy, such as a cytokine therapy, a T-cell therapy, an NK therapy,
an immune system
checkpoint inhibitor (e.g. an antibody), chemotherapy, radiotherapy,
immunostimulating
substances, gene therapy, and dendritic cells.
10. The method of claim 9 wherein the additional cancer therapy is one or
more of Actimide,
Azacitidine, Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin,
Chlorambucil,
Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine,
Doxorubicin,
Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea,
Idarubicin,
Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan,
Mercaptopurine,
Methotrexate, Mitoxantrone, Nivolumab, Oxaliplatin, Paclitaxel, Pembrolizumab,
Pemetrexed,
Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine,
Vincristine,
Vindesine and Vinorelbine.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
1
IMMUNOGENIC ARGINASE PEPTIDES
Technical field
The present invention relates to novel peptide compounds, such as fragments of
Arginase
1, as well as compositions, uses, and kit-of-parts comprising these peptide
compounds.
Furthermore, the invention concerns nucleic acids, vectors, and host cells
expressing said peptide
compounds, for use in a method for treatment or prevention of a cancer, either
alone or when
administered simultaneously or sequentially with an additional cancer therapy.
Background Art
Arginase is an enzyme that catalyses a reaction which converts the amino acid
L-arginine
into L-omithine and urea. This depletes the microenvironment of arginine and
leads to a
suppression of tumor- specific cytotoxic T-cell responses. Increased Arginase
activity has been
detected in the cancer cells of patients with breast, lung, colon or prostate
cancer [1]. It has been
.. shown both in vitro and in vivo that mouse macrophages transfected with a
rat Arginase gen
promote the proliferation of co-cultured tumour cells [2]. Furthermore
induction of Arginase
expression by macrophages has been shown to increase tumour vascularization
through
polyamine synthesis. The results of a murine lung carcinoma model showed that
there existed a
subpopulation of mature tumor-associated myeloid cells that expressed high
levels of Arginase.
These tumor-associated myeloid cells depleted the extracellular L-Arginine
which inhibited
antigen-specific proliferation of the tumor infiltrating lymphocytes (TILs).
Injection of an
Arginase inhibitor blocked the growth of the lung carcinoma in the mice. This
shows how
induction of Arginase expression in tumor cells and tumor associated myeoloid
cells might
promote tumor growth by suppression of the anti- tumor immune responses
through negative
effects on TILs.
MDSCs (myeloid-derived suppressor cells) inhibit the activation,
proliferation, and
cytotoxicity of effector T cells and natural killer cells, as well as induce
Treg differentiation and
expansion. Both cancer cells and MDSCs can suppress T cells by manipulating L-
arginine
metabolism via the enzymes nitric-oxide synthase (NOS) and arginase. Many
tumours exhibit
increased expressions of arginase and inducible NOS (iNOS), leading to
arginine depletion from

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
2
the tumour microenvironment [1]. Several studies emphasize the importance of
this altered
tumour arginine metabolism in the suppression of tumour-specific T-cell
responses, and it was
recently demonstrated that AML blasts show an arginase-dependent ability to
inhibit T-cell
proliferation and hematopoietic stem cells. Furthermore, arginase and iNOS
inhibitors reduce the
suppressive activity of AML [2].
Summary of the invention
The present inventors have identified new immunogenic epitopes from Arginase
1.
Furthermore, peripheral blood mononuclear cells (PBMC) from melanoma patients
have been
analysed for the presence of specific T-cell responses against Arginase 1-
derived peptides and
strong immune responses against the new immunogenic epitopes were detected.
Moreover,
frequent immune responses were detected against several peptide fragments. It
has also been
shown that the immune responses towards Arginase 1 indeed were mediated by CD4
and CD8 T
cells, and that both CD8 and CD4 T cells can recognize Arginase 1 derived
peptides on the
surface of target cells.
The development of novel immune therapies for cancer requires a thorough
understanding of the molecules that are involved in the pathogenesis as well
as the specific
proteins recognized by the immune system. In the clinical setting the
induction of Arginase
specific immune responses could in addition to the killing of cancer cells
support anti-cancer
immune responses in general by suppressing the immune suppressive function of
Arginase
expressing cells especially MDSC and tumor-associated macrophages (TAMs).
Hence, since
Arginase-expressing cells antagonize the desired effects of other
immunotherapeutic approaches
targeting myeloid dendritic cells e.g. by vaccination, would consequently be
highly synergistic
with additional anti-cancer immunotherapy.
Provided herein is an isolated, immunogenic polypeptide fragment of a human
Arginase
protein of SEQ ID NO: 1 (Arginase 1) or SEQ ID NO: 60 (Arginase 2). The
fragment is typically
up to 8, 9, 10, 15, 20, 25, 30, 45, 50 or 55 amino acids in length. The
fragment may comprise or
consists of a sequence of at least 8, 9, 10, 20, 30, 40 or 50 consecutive
amino acids of either of
SEQ ID NO: 52 or SEQ ID NO: 58. The polypeptide fragment may comprise or
consist of the
amino acid sequence of any one of SEQ ID NOs: 52, 50, 51, 34, 35, 36, 37, 9,
53, 54, 2 to 33, or
38 to 49.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
3
The polypeptide fragment may have one or more of the following additional
features:
a. the C terminal amino acid is replaced with the corresponding amide; and/or
b. the L at the position corresponding to position 190 of SEQ ID NO: 1 is
replaced with
I; and/or
c. the R at the position corresponding to position 205 of SEQ ID NO: 1 is
replaced with
K; and/or
d. at least one additional moiety is attached to the N and/or C terminus,
optionally
wherein said additional moiety is a hydrophilic amino acid such as R or K;
and/or
e. lacks or has reduced arginase activity relative to the corresponding full
length
arginase.
Also provided is a composition comprising a said fragment, a pharmaceutically
acceptable
diluent or carrier, and optionally an adjuvant. Said composition or said
fragment may be for use
in a method of treating or preventing a disease or condition such as cancer,
or for use in the
manufacture of a medicament for treating or preventing a disease or condition
such as cancer.
Said composition may optionally be described as a vaccine. Also provided is a
method of
treating or preventing a disease or condition such as cancer, the method
comprising
administering said composition or said fragment to a subject in need thereof.
The polypeptide fragment may be interchangeably described herein as a peptide
compound or a peptide.
In one aspect the present disclosure concerns a peptide compound of Arginase 1
selected
from:
a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of
from 8 to 321
amino acids,
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 1 or
.. the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 1 or the peptide fragment of a),
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide;
or a pharmaceutically acceptable salt thereof In the above-mentioned context,
"functional"
means "capable of stimulating an immune response to the Arginase of SEQ ID NO:
F. The
homologue b) and analogue c) preferably have reduced (or zero) arginase
function.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
4
In an embodiment the peptide compound is selected from a) a peptide fragment
of SEQ
ID NO 1 consisting of a consecutive sequence of from 8 to 321 amino acids,
wherein the C-
terminal amino acid also comprises the amide; or a pharmaceutically acceptable
salt thereof. In a
further embodiment the peptide fragment consists of a consecutive sequence in
the range of from
8 to 300 amino acids, 8 to 250 amino acids, 8 to 200 amino acids, 8 to 150
amino acids, 8 to 120
amino acids, e.g. 10 to 100 amino acids, 20 to 80 ami-no acids, 30 to 60 amino
acids, 40 to 50
amino acids. In a still further embodiment the peptide fragment of SEQ ID NO 1
is selected from
the group consisting of ARG(1-310), ARG(1-301), ARG(1-291), ARG(11-322),
ARG(21-322),
ARG(30-322), ARG(40-322), ARG(11-310), ARG(11-301), ARG(11-291), ARG(21 -310),
ARG(21-301), ARG(21-291), ARG(30-310), ARG(30-301), ARG(30-291), ARG(40-310),
ARG(40-301), and ARG(40-291).
In a further embodiment the arginase activity is reduced compared to Arginase
1 as
measured by an arginase activity assay. In one embodiment the arginase
activity is reduced to
inactivity. In another embodiment the arginase activity assay is selected from
the Arginase
Activity Colorimetric Assay Kit ( BioVision Arginase assay # K755-100).
In a still further embodiment the consecutive sequence comprises one or more
sequences
selected from any one of SEQ ID NOs: 52, 50, 51, 37, 36, 35, 34, 9, 53, 54, 2
to 33, or 38 to 39
. In one embodiment the consecutive sequence comprises the sequence selected
from SEQ ID
NO 52, 50, 51, 37, 36, 35, 34, 9, 53 or 54.
In a further embodiment the peptide fragment under a), the functional
homologue under
b), or the functional analogue under c) is functional in the sense that it
activates T cells that
recognizes Arginase 1 expressing cells. In one embodiment the activation is
determined by the
ELISPOT assay described herein.
Also disclosed herein is a nucleic acid encoding the peptide compound of the
present
.. invention. The peptide compound of the present invention is selected from
any one of the above
embodiments. In one embodiment the nucleic acid is selected from the group
consisting of DNA
and RNA.
Also disclosed herein is a vector comprising the nucleic acid of the present
invention.
The nucleic acid of the present invention is selected from any one of the
above embodiments,
and the peptide compound of the present invention is selected from any one of
the above
embodiments. In one embodiment the vector is selected from a virus vector.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
In a further aspect the present disclosure relates to a host cell comprising
the vector of the
present invention. The vector is selected from any one of the above
embodiments, the nucleic
acid is selected from any one of the above embodiments, and the peptide
compound is selected
from any one of the above embodiments. In one embodiment the host cell is
selected from a
5 mammalian cell.
The vector preferably comprises a nucleic acid encoding an inactive sequence
of
Arginase 1 , that is a sequence which lacks arginase function.
Any peptide, nucleic acid, vector, or host cell described herein may be
provided in a
composition, optionally together with a pharmaceutically acceptable additive,
such as a carrier or
diluent. The composition may optionally also comprise an adjuvant. The
composition may be
for use as a medicament. The composition may be for use in the manufacture of
a medicament
for treating or preventing a disease.
The composition may be for use in a method for treatment or prevention of a
disease,
disorder or condition selected from cancer. In one embodiment the cancer is a
tumor forming
cancer disease. The adjuvant may be selected from the group consisting of
bacterial DNA based
adjuvants, oil/surfactant based adjuvants, viral dsRNA based adjuvants,
imidazochinilines, and a
Montanide ISA adjuvant.
A method of treatment or prevention of a disease as disclosed herein may
comprise
administering to a subject an effective amount of:
a) a composition as described above, and
b) a composition comprising at least one second active ingredient, selected
from an
immunostimulating compound, such as an interleukin, e.g. IL-2 and or IL-21, an
anti-cancer
agent, such as a chemotherapeutic agent, e.g. Actimide, Azacitidine, Azathio-
prine, Bleomycin,
Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophospha-mide,
Cytarabine,
Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide,
Fludarabine,
Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotec-an, Lenalidomide,
Leucovorin,
Mechlorethamine, Melphalan, Mercaptopurine, Metho-trexate, Mitoxantrone,
Oxaliplatin,
Paclitaxel, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine,
Valrubicin,
Vinblastine, Vincristine, Vindesine and Vinorelbine, or a checkpoint
inhibitor, e.g. antibody such
as nivolumab or pembrolizumab.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
6
In The provided compositions may be administered simultaneously or
sequentially. The
compositions may be provided as components in a kit-of-parts.
In a further aspect the present disclosure relates to a method of treating a
clinical
condition characterized by expression of Arginase 1 of SEQ ID NO 1, the method
comprising
administering to an individual suffering from said clinical condition an
effective amount of the
peptide, the nucleic acid, the vector or the host cell described above.
In a further aspect the present disclosure relates to a method of stimulation
of arginase 1
specific T-cells, such as CD4 and CD8 T-cells, in a cancer patient, the method
comprising
administering to the cancer patient an effective amount of the peptide
compound, or the nucleic
.. acid, or the vector, or the host cell as described above.
In a still further aspect the present disclosure relates to a method of
suppressing an
immune suppressive function of Arginase 1 expressing cells, in a cancer
patient, the method
comprising administering to the cancer patient an effective amount of the
peptide compound, or
the nucleic acid, or the vector, or the host cell as described above.
In a further aspect the present disclosure relates to use of the peptide
compound, or the
nucleic acid, or the vector, or the host cell, as described above for the
manufacture of a
medicament, such as an immunotherapeutic composition or vaccine, for the
treatment or
prevention of a cancer, wherein said cancer is optionally characterized by
expression of Arginase
1.
In a still further aspect the present disclosure relates to the peptide
compound, or the
nucleic acid, or the vector, or the host cell, for use in a method for
treatment or prevention of a
cancer, optionally when administered simultaneously or sequentially with an
additional cancer
therapy.
The additional cancer therapy may be selected from the group consisting of a
cytokine
therapy, a T-cell therapy, an NK therapy, an immune system checkpoint
inhibitor, chemotherapy,
radiotherapy, immunostimulating substances, gene therapy, antibodies and
dendritic cells. In one
embodiment the additional cancer therapy is selected from an immune system
checkpoint
inhibitor, such as a checkpoint blocking antibody (e.g. selected from
nivolumab or
pemrbolizumab), or is selected from the group consisting of Actimide,
Azacitidine,
.. Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin,
Chlorambucil,
Cyclophosphamide, Cytarabine, Dauno-rubicin, Docetaxel, Doxifluridine,
Doxorubicin,

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
7
Epirubicin, Etoposide, Fludarabine, Fluor-ouracil, Gemcitabine, Hydroxyurea,
Idarubicin,
Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan,
Mercaptopurine,
Methotrexate, Mitoxantroneõ Oxaliplatin, Paclitaxelõ Pemetrexed, Revlimid,
Temozolomide,
Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and
Vinorelbine.
Brief description of drawings
Figure 1 shows T cell responses in samples from 6 cancer patients to the
peptides consisting of
each of the amino acids sequences of SEQ ID NOs: 2 to 16 (Argl to Arg15).
Figure 2 shows T cell responses in samples from 6 cancer patients against the
peptide consisting
of each of the amino acid sequence of SEQ ID NO: 9 (Arg8).
Figure 3 shows T cell responses in samples from 3 cancer patients to the
peptides of consisting
of each of the amino acid sequences of SEQ ID NOs: 18 to 48 (Argl-1 to Arg1-
31).
Figure 4 shows T cell responses in samples from 8 healthy individuals against
the peptides
consisting of the amino acid sequence of each of SEQ ID NO: 9 (Arg8), SEQ ID
NO: 21 (Argl -
4), SEQ ID NO: 29 (Argl -12), SEQ ID NO: 34 (Argl -17), SEQ ID NO: 35 (Argl -
18), SEQ ID
NO: 36 (Argl -19), SEQ ID NO: 37 (Argl -20), SEQ ID NO: 40 (Argl -23), SEQ ID
NO: 44
(Argl-17).
Figure 5 shows CD8 positive culture can kill target cells loaded with the
peptide consisting of
each of the amino acid sequence of SEQ ID NO: 9 (Arg8).
Figure 6 shows killing of Arginase positive cancer (melanoma) cell lines FM3
and FM93 by
Arginase specific T cells.
Figure 7 shows flow cytometric analysis of CD4 Tcells from a cancer patient,
assessed by
intracellular staining for IFN-g (PE-Cy7A) and TNF-alpha (APC-A) following
culture either
without (top panel) or with (bottom panel) stimulation with the peptide
consisting of each of the
amino acid sequence of SEQ ID NO: 9 (Arg8). Similar results are shown in
Figure 8.
Figure 9 shows that multiple arginase-1 peptides are recognized by PBMCs from
3 melanoma
patients (9A) and 8 healthy donors (9B) when assessed by IFNy ELISPOT.
Peptides are
described by reference to the start and end positions of their sequences
within the sequence of
human Arginase 1. Spot counts are given as a difference between averages of
the wells
stimulated with the peptide and control wells. Peptide and control
stimulations were performed
in duplicates or triplicates.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
8
Figure 10 shows that the region corresponding to positions 161 to 210 of SEQ
ID NO: 1 is
widely recognized by cancer patient and healthy donor PBMCs
A ¨ shows the proportion of CD4+ T cells in tumour infiltrating lymphocytes
(TILs) from
melanoma patient which release IFNy in response to the peptides consisting of
the amino acid
sequences of each of SEQ ID NO: 34 (Arg161-180), SEQ ID NO: 35 (Arg171-190)
and SEQ ID
NO: 36 (Arg181-200). Peptide names refer to the start and end positions of
their sequences
within the sequence of human Arginase 1
B ¨ Left: Responses against Arg(161-190) peptide in PBMCs from 5 selected
cancer patients
and four healthy donors. Right: Exemplary ELISPOT wells for responses against
Arg(161-190)
peptide or control in 2 healthy donors (HD..) and 2 cancer patients (MM..).
C ¨ Left: Responses against Arg(181-210) peptide in PBMCs from 5 selected
cancer patients
and four healthy donors. Right: Exemplary ELISPOT wells for responses against
Arg(161-190)
peptide in 2 healthy donors (HD..) and 2 cancer patients (MM..).
Spot counts are given as a difference between averages of the wells stimulated
with the peptide
and control wells. Peptide and control stimulations were performed in
triplicates.
Figure 11 shows further analysis of responses to the region corresponding to
positions 161 to 210
of SEQ ID NO: 1
A ¨ Top shows exemplary flow cytometry analysis of CD4 Tcells from a cancer
patient, assessed
by intracellular staining for IFN-g (PE-Cy7A) and TNF-alpha (APC-A) release
following 8 hour
stimulation with the peptide Arg(161-190) or unstimulated without peptide.
Bottom shows the
proportion TNF-a releasing CD4+ T cells in this assay for 2 healthy donors
(HD..) and 1 cancer
patient (MM..).
B ¨ summary of IFNy ELISPOT results following stimulation of PBMCs from 3
cancer patients
with the peptide Arg(161-190) alone, or with antibody blocking of expression
of HLA Class I
(anti-Class I; W6/32) or HLA Class II (anti-Class II; Tii39). Spot counts are
given as a difference
between the wells stimulated with the peptide and control wells. Peptide and
control stimulations
were performed in duplicates or triplicates.
C ¨ Left: IFNy ELISPOT responses from Arginase specific CD4 T cells (produced
by repeated
stimulation with the peptide consisting of the amino acid sequence of SEQ ID
NO: 9 (ArgShort))
when stimulated with ArgShort, the 30mer consisting of the sequence from
positions 161 to 190
of Arginase 1 (Arg 161-190), or the 50mer consisting of the sequence from
positions 161 to 210

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
9
of Arginase 1 (Arg 161-210), or when unstimulated (control). Right: Exemplary
flow cytometric
analysis of the Arginase specific CD4 T cells assessed by intracellular
staining for IFN-g (PE-
Cy7A) and TNF-alpha (APC-A) release following 8 hour stimulation with the
peptide Arg(161-
190) or control.
Figure 12 shows Arginase-specific T cells recognize arginase-expressing immune
cells
A and C ¨ IFNy response by the arginase specific T cell cultures from two
different melanoma
patients (MM01 and MM05) to autologous dendritic cells electroporated and
trasnfected with
irrelevant control mRNA (DC Mock) or Arginase-1 mRNA (DC Arg mRNA). Effector
to target
ratio 10:1.
B and D ¨ IFNy response by the arginase specific T cell cultures from two
different melanoma
patients to autologous B cells electroporated and transfected with irrelevant
control mRNA (DC
Mock) or Arginase-1 mRNA (DC Arg mRNA) Effector to target 2:1.
E ¨ Bottom: IFNy response by arginase specific T cell culture towards
autologous dendritic cells
electroporated and transfected with irrelevant control mRNA (DC Mock),
arginase mRNA (DC
Arg mRNA) or arginase mRNA containing DC-LAMP signal sequence (DC LAMP Arg
mRNA). Top: representative ELISPOT well images. Control and transfected cell
stimulations
were performed in duplicates or triplicates.
Figure 13 is a sequence alignment of human Arginase 1 and murine Arginase 1.
Figure 14 shows that arginase specific immune responses are increased in
C57BL/6 mice
following vaccination with different peptides disclosed herein.
Figure 15 shows that arginase specific immune responses are increased in
Balb/c mice following
vaccination with different peptides disclosed herein.
Figure 16 shows reduction in tumour volumes in a mouse model of cancer
vaccination with
different peptides disclosed herein.
Brief description of Sequences
SEQ ID NO: 1 is the wild type amino acid sequence of human Arginase 1
SEQ ID NOS: 2 to 54 are the amino acid sequences of peptide fragments of human
Arginase 1
SEQ ID NOs: 55 to 57 are the amino acid sequences of peptide fragments of
murine Arginase 1
SEQ ID NOs: 58 is the amino acid sequence of a peptide fragment of human
Arginase 2
SEQ ID NO: 59 is the wild type amino acid sequence of murine Arginase 1

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
SEQ ID NO: 60 is the wild type amino acid sequence of human Arginase 2
SEQ ID NO: 61 is an alternative sequence of human Arginase 1
Detailed Description of the invention
5 The present invention is concerned with immunogenic polypeptide
fragments of Arginase
proteins. Such fragments may be useful as vaccines. By "immunogenic" it is
meant that a
polypeptide fragment is capable of eliciting an immune response, preferably a
T-cell response, in
at least one individual after administration to said individual. A polypeptide
may be identified as
immunogenic using any suitable method, including in vitro methods. For
example, a peptide may
10 be identified as immunogenic if it has at least one of the following
characteristics:
(0 It is capable of eliciting IFN-y -producing cells in a PBL
population of at least one
cancer patient as determined by an ELISPOT assay, and/or
(ii) It is capable of in situ detection in a sample of tumor tissue of
CTLs that are reactive
with the corresponding arginase; and/or
(iii) It is capable of inducing the in vitro growth of specific T-cells.
Methods suitable for determining whether a polypeptide is immunogenic active
are also provided
in the Examples section below.
The inventors have determined that multiple regions of the Arginase 1 sequence
of SEQ
ID NO: 1 are immunogenic. These include SEQ ID NOs: 52, 50, 51, 37, 36, 35,
34, 9, 53, 54, 2
to 33, or 38 to 49. SEQ ID NO: 52 corresponds to the region of Arginasel from
position 161 to
210 of SEQ ID NO: 1. This region may be described as a hotspot for
immunogenicity because it
comprises sequences against which T cell responses are detected most
frequently amongst cancer
patients and healthy subjects. The immunogenic polypeptide may be upto 50 or
55 amino acids
in length and/or comprise at least 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or all
50 consecutive amino
acids of SEQ ID NO: 52. Said consecutive amino acids may preferably comprise
or consist of
any one of SEQ ID NOs: 50, 51, 34, 35, 36, 37, 9, 53 and 54. Each of SEQ ID
NOs: 50, 51, 34,
35, 36, 37, 9, 53 and 54 comprises sequences from within the sequence of SEQ
ID NO: 52.
Specifically, each of SEQ ID NOs: 50, 51, 34, 35, 36, 37, 9, 53 and 54
corresponds to the regions
of SEQ ID NO: 1 defined by positions 161-190, 181-210, 161-180, 171-190, 181-
200, 191-210,
174-182, 172-179 and 193-200, respectively.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
11
The immunogenic polypeptide fragment may be upto 30, 35, 40, 45, 50 or 55
amino acids
in length and comprise at least 8, 9, 10, 15, 20, 25 or all 30 consecutive
amino acids of SEQ ID
NO: 50 or 51. The immunogenic polypeptide fragment may be upto 20, 25, 30, 35,
40, 45, 50 or
55 amino acids in length and comprise at least 8, 9, 10, 15 or all 20
consecutive amino acids of
.. any one of SEQ ID NOs: 37, 36, 35, 34.
The polypeptide fragment preferably comprises at least 8, 9, 10, 15 or all 20
consecutive
amino acids from the region defined by positions 181-200 of SEQ ID NO: 1 (that
is SEQ ID NO:
36) or most preferably at least 8, 9, 10, 15 or all 20 consecutive amino acids
from the region
defined by positions 191-200 of SEQ ID NO: 1 (that is SEQ ID NO: 37). The
polypeptide
.. fragment may comprise at least 8, 9, 10, 15 or all 20 consecutive amino
acids from the region
defined by positions 161-180 of SEQ ID NO: 1 (that is SEQ ID NO: 34) or
positions 171-190 of
SEQ ID NO: 1 (that is SEQ ID NO: 35). In the former (SEQ ID NO: 34), the
cysteine
corresponding to position 168 may optionally be replaced by conservative
substitution, e.g. to
improve solubility or stability.
The immunogenic polypeptide fragment may be upto 8 or 9 amino acids in length
and
may comprise at least 8 or 9 consecutive amino acids of any one of SEQ ID NOs:
9, 53 or 54.
The immunogenic polypeptide fragment may comprise or consist of the amino acid
sequence of
any one of SEQ ID NOs: 52, 50, 51, 34, 35, 36, 37, 9, 53, 54, 2 to 33, or 38
to 49.
The inventors also identified responses in sequences which correspond to other
regions of
.. SEQ ID NO: 1, including the regions defined by (or at least partially
overlapping with) positions
221-240 (see SEQ ID NOs: 40,7, 14); 271-290 (see SEQ ID NOs: 45,8, 10, 11,
12); 111-130
(see SEQ ID NOs: 29); 1-20 (see SEQ ID NOs: 18); 61-80 (see SEQ ID NOs: 24);
131-150 (see
SEQ ID NOs: 31, 4); 141-160 (see SEQ ID NOs: 32, 4); 51-70 (see SEQ ID NOs:
23); 151-170
(see SEQ ID NOs: 33, 4); 211-240 (see SEQ ID NOs: 39, 6); and 281-300 (see SEQ
ID NOs:
46). The immunogenic polypeptide fragment may thus comprise at least 8, 9 or
more
consecutive amino acids from any one of these regions or sequences. The
immunogenic
polypeptide fragment may comprise or consist of the amino acid sequence of any
one of these
regions or sequences.
Human Arginase 2 is related to Arginase 1 and there are some similarities in
sequence
.. (see SEQ ID NO: 60 versus SEQ ID NO: 1, respectively). The regions of
interest in Arginase 1
are thus also likely to be of interest in Arginase 2. Accordingly, any one of
the sequences

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
12
described in the preceding section may have any one or more amino acids
replaced with the one
or more amino acids in the corresponding positions in Arginase 2. The entirety
of the said
sequence may be replaced with the corresponding sequence in Arginase 2. The
immunogenic
polypeptide may comprise or consist of a sequence of SEQ ID NO: 60 which is
defined by the
positions in SEQ ID NO: 60 that correspond to the positions in SEQ ID NO: 1 of
any one of SEQ
ID NOs: 52, 50, 51, 34, 35, 36, 37, 9, 53, 54, 2 to 33, or 38 to 49. For
example, the hotspot
region defined by positions 161-210 of SEQ ID NO: 1 corresponds to positions
180-229 of SEQ
ID NO: 60 which are represented herein as SEQ ID NO: 58. Thus, references to
SEQ ID NO: 52
may be replaced by references to SEQ ID NO: 58. The immunogenic polypeptide
may comprise
at least 8, 9, or preferably at least 20 or 30 consecutive amino acids of SEQ
ID NO: 58.
Human Arginase 1 is also highly similar to murine Arginase 1 (see SEQ ID NO:
59
versus SEQ ID NO: 1, respectively, plus Figure 13). Accordingly, any one of
the sequences
described in the preceding section for human Arginase 1 may have any one or
more amino acids
replaced with the one or more amino acids in the corresponding positions in
murine Arginase 1.
The entirety of the said sequence may be replaced with the corresponding
sequence in murine
Arginase 1. The immunogenic polypeptide may comprise or consist of a sequence
of SEQ ID
NO: 59 which is defined by the positions in SEQ ID NO: 59 that correspond to
the positions in
SEQ ID NO: 1 of any one of SEQ ID NOs: 52, 50, 51, 34, 35, 36, 37, 9, 53, 54,
2 to 33, or 38 to
49. For example, the hotspot region defined by positions 161-210 of SEQ ID NO:
1
corresponds to positions 161-210 of SEQ ID NO: 59, which are represented
herein as SEQ ID
NO: 57. Thus, references to SEQ ID NO: 52 may be replaced by references to SEQ
ID NO: 57.
The immunogenic polypeptide may comprise at least 8, 9, or preferably at least
20 or 30
consecutive amino acids of SEQ ID NO: 57. In effect, this means that the L at
the position
corresponding to position 190 of SEQ ID NO: 1 may be replaced with I and/or
that the R at the
position corresponding to position 205 of SEQ ID NO: 1 is replaced with K, in
any polypeptide
fragment described herein which encompasses those residues.
In any polypeptide fragment described herein, the C terminal amino acid may
optionally
be replaced with the corresponding amide, to improve solubility and/or to aid
with manufacture /
isolation. Similarly, the polypeptide may have attached at the N and/or C
terminus at least one
additional moiety to improve solubility and/or to aid with manufacture /
isolation. Suitable

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
13
moieties include hydrophilic amino acids. For example, the amino acids KR may
be added at the
N terminus and/or the amino acids RK may be added in order at the C terminus.
Any polypeptide fragment described herein preferably has reduced arginase
activity
relative to the corresponding full-length arginase. A reduction in arginase
activity may include
the reduction to inactivity. A suitable assay for arginase activity is the
Arginase Activity
Colorimetric Assay Kit (B ioVi s ion Arginase assay # K755-100).
In one aspect the present disclosure concerns a peptide compound of Arginase 1
selected
from:
a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of
from 8 to 321
amino acids,
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 1 or
the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 1 or the peptide fragment of a),
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide;
or a pharmaceutically acceptable salt thereof
In an embodiment the peptide compound is selected from b) a functional
homologue
having at least 70%, 80%, 90%, or 95% identity to SEQ ID NO 1 or the peptide
fragment of a),
wherein the C-terminal amino acid also comprises the amide; or a
pharmaceutically acceptable
salt thereof. In one embodiment the functional homologue has at least 80%
identity to SEQ ID
NO 1. In a further embodiment the functional homologue has at least 90%
identity to SEQ ID
NO 1. In a further embodiment the functional homologue has at least 95%
identity to SEQ ID
NO 1. In a further embodiment the functional homologue has at least 70%
identity to the peptide
fragment of a). In a further embodiment the functional homologue has at least
80% identity to
the peptide fragment of a). In a further embodiment the functional homologue
has at least 90%
identity to the peptide fragment of a). In a further embodiment the functional
homologue has at
least 95% identity to the peptide fragment of a).
In another embodiment the peptide compound is selected from c) a functional
analogue
wherein at least one amino acid has been deleted, inserted and/or substituted
in SEQ ID NO 1 or
the peptide fragment of a), wherein the C-terminal amino acid also comprises
the amide; or a
pharmaceutically acceptable salt thereof.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
14
In a further embodiment the peptide compound is selected from a) a peptide
fragment of
SEQ ID NO 1 consisting of a consecutive sequence of from 8 to 321 amino acids,
wherein the C-
terminal amino acid also comprises the amide; or a pharmaceutically acceptable
salt thereof. In a
further embodiment the peptide fragment consists of a consecutive sequence in
the range of from
.. 8 to 300 amino acids. In a further embodiment the peptide fragment consists
of a consecutive
sequence in the range of from 8 to 250 amino acids. In a further embodiment
the peptide
fragment consists of a consecutive sequence in the range of from 8 to 200
amino acids. In a
further embodiment the peptide fragment consists of a consecutive sequence in
the range of from
8 to 150 amino acids. In a further embodiment the peptide fragment consists of
a consecutive
.. sequence in the range of from 8 to 120 amino acids. In a further embodiment
the peptide
fragment consists of a consecutive sequence in the range of from 10 to 100
amino acids. In a
further embodiment the peptide fragment consists of a consecutive sequence in
the range of from
to 80 amino acids. In a further embodiment the peptide fragment consists of a
consecutive
sequence in the range of from 30 to 60 amino acids. In a further embodiment
the peptide
15 fragment consists of a consecutive sequence in the range of from 40 to
50 amino acids. In a still
further embodiment the peptide fragment of SEQ ID NO 1 is selected from the
group consisting
of ARG(1-310), ARG(1-301), ARG(1-291), ARG(11-322), ARG(21-322), ARG(30-322),
ARG(40-322), ARG(11-310), ARG(11-301), ARG(11-291), ARG(21-310), ARG(21-301),
ARG(21-291), ARG(30-310), ARG(30-301), ARG(30-291), ARG(40-310), ARG(40-301),
and
20 .. ARG(40-291). In a still further embodiment the peptide fragment of SEQ
ID NO 1 is selected
from ARG(1-310). In a still further embodiment the peptide fragment of SEQ ID
NO 1 is
selected from ARG(1-301). In a still further embodiment the peptide fragment
of SEQ ID NO 1
is selected from ARG(1-291). In a still further embodiment the peptide
fragment of SEQ ID NO
1 is selected from ARG(11-322). In a still further embodiment the peptide
fragment of SEQ ID
.. NO 1 is selected from ARG(21-322). In a still further embodiment the
peptide fragment of SEQ
ID NO 1 is selected from ARG(30-322). In a still further embodiment the
peptide fragment of
SEQ ID NO 1 is selected from ARG(40-322). In a still further embodiment the
peptide fragment
of SEQ ID NO 1 is selected from ARG(11-310). In a still further embodiment the
peptide
fragment of SEQ ID NO 1 is selected from ARG(11-301). In a still further
embodiment the
.. peptide fragment of SEQ ID NO 1 is selected from ARG(11-291). In a still
further embodiment
the peptide fragment of SEQ ID NO 1 is selected from ARG(21-310). In a still
further

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
embodiment the peptide fragment of SEQ ID NO 1 is selected from ARG(21-301).
In a still
further embodiment the peptide fragment of SEQ ID NO 1 is selected from ARG(21-
291). In a
still further embodiment the peptide fragment of SEQ ID NO 1 is selected from
ARG(30-310). In
a still further embodiment the peptide fragment of SEQ ID NO 1 is selected
from ARG(30-301).
5 In a still further embodiment the peptide fragment of SEQ ID NO 1 is
selected from ARG(30-
291). In a still further embodiment the peptide fragment of SEQ ID NO 1 is
selected from
ARG(40-310). In a still further embodiment the peptide fragment of SEQ ID NO 1
is selected
from ARG(40-301). In a still further embodiment the peptide fragment of SEQ ID
NO 1 is
selected from ARG(40-291). In a still further embodiment the peptide fragment
of SEQ ID NO 1
10 is selected from ARG(161-190). In a still further embodiment the peptide
fragment of SEQ ID
NO 1 is selected from ARG(181-210). In a still further embodiment the peptide
fragment of SEQ
ID NO 1 is selected from ARG(161-210).
It is to be understood that when the peptide fragment consists of a
consecutive sequence
in the range of from 8 to 120, it may at the same time be selected within the
sequence of for
15 instance ARG(40-291), whereas a peptide fragment consisting of a
consecutive sequence in the
range of from 8 to 321, cannot be at the same time be selected within the
sequence of for
instance ARG(40-291), this is known to the person skilled in the art.
Otherwise all combinations
are contemplated within the present invention.
It is also to be understood that ARG(x-y), wherein x and y are integers
selected from 1-
322 as used herein means a peptide fragment of Arginase 1 having the SEQ ID NO
1 as defined
herein, wherein x is the N-terminal amino acid and y is the C-terminal amino
acid, for instance
ARG(174-182) indicates the peptide fragment from amino acid 174 of SEQ ID NO 1
to amino
acid 182 of SEQ ID NO 1 wherein amino acid 174 is I and amino acid 182 is V.
The term "identity" as used herein refers to a relationship between the
sequences of two
or more peptides, such as polypeptides, as determined by comparing the
sequences. In the art,
"identity" also means the degree of sequence relatedness between proteins or
polypeptides, as
determined by the number of matches between strings of two or more amino acid
residues.
"Identity" measures the percent of identical matches between the smaller of
two or more
sequences with gap alignments (if any) addressed by a particular mathematical
model or
computer program (i.e., "algorithms"). Identity of related proteins or
peptides can be readily
calculated by known methods. Such methods include, but are not limited to,
those described in

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
16
Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press,
New York, 1988;
Biocomputing: Informatics and Genome Pro-jects, Smith, D. W., ed., Academic
Press, New
York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and
Griffin, H. G.,
eds., Humana Press, New Jersey, 1994; Se-quence Analysis in Molecular Biology,
von Heinje,
G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux,
J., eds., M.
Stockton Press, New York, 1991; and Carillo et al., SIAM J. Applied Math., 48,
1073, (1988).
Preferred methods to determine identity are designed to give the largest match
between
the sequences tested. Methods to determine identity are described in publicly
available computer
programs. Preferred computer program methods to determine identity between two
sequences
include the GCG program package, including GAP (Devereux et al., Nucl. Acid.
Res., 12, 387,
(1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.),
BLASTP,
BLASTN, and FASTA (Altschul et al., J. Mol. Biol., 215, 403-410, (1990)). The
BLASTX
program is publicly available from the National Center for Biotechnology
Information (NCBI)
and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md.
20894;
Altschul et al., supra). The well known Smith Waterman algorithm may also be
used to
determine identity.
For example, using the computer algorithm GAP (Genetics Computer Group,
University
of Wisconsin, Madison, Wis.), two proteins for which the percent sequence
identity is to be
determined are aligned for optimal matching of their respective amino acids
(the "matched span",
.. as determined by the algorithm). A gap opening penalty (which is calculated
as 3 times the
average diagonal; the "average diagonal" is the average of the diagonal of the
comparison matrix
being used; the "diagonal" is the score or number assigned to each perfect
amino acid match by
the particular comparison matrix) and a gap extension penalty (which is
usually {fraction (1/10)}
times the gap opening penalty), as well as a comparison matrix such as PAM 250
or BLOSUM
62 are used in conjunction with the algorithm. A standard comparison matrix
(see Dayhoff et al.,
Atlas of Protein Sequence and Structure, vol. 5, supp.3 (1978) for the PAM 250
comparison
matrix; Henikoff et al., Proc. Natl. Acad. Sci USA, 89, 10915-10919, (1992)
for the BLOSUM
62 comparison matrix) is also used by the algorithm. Preferred parameters for
a protein or
peptide sequence comparison include the following: Algorithm: Needleman et
al., J. Mol. Biol,
48, 443-453, (1970); Comparison matrix: BLOSUM 62 from Henikoff et al., Proc.
Natl. Acad.
Sci. USA, 89, 10915-10919, (1992); Gap Penalty: 12, Gap Length Penalty: 4,
Threshold of

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
17
Similarity: 0. The GAP program is useful with the above parameters. The
aforementioned
parameters are the default parameters for protein comparisons (along with no
penalty for end
gaps) using the GAP algorithm.
In a further embodiment the arginase activity is reduced compared to Arginase
1 as
measured by an arginase activity assay. In one embodiment the arginase
activity is reduced to
inactivity. In another embodiment the arginase activity assay is selected from
the Arginase
Activity Colorimetric Assay Kit ( BioVision Arginase assay # K755-100). In a
further
embodiment the arginase activity is reduced to inactivity compared to Arginase
1 as measured by
the arginase activity assay is selected from the Arginase Activity
Colorimetric Assay Kit.
In a still further embodiment the consecutive sequence comprises one or more
sequences
selected from any one of SEQ ID NO 2-17. In one embodiment the consecutive
sequence
comprises the sequence selected from SEQ ID NO 9. In another embodiment the
consecutive
sequence comprises only the sequence selected from SEQ ID NO 9, but not any
one of the SEQ
ID NO 2-8 or 10-17.
In a further embodiment the consecutive sequence comprises the sequence having
SEQ
ID NO 50. In a still further embodiment the consecutive sequence comprises the
sequence
having SEQ ID NO 51. In a further embodiment the consecutive sequence
comprises the
sequence having SEQ ID NO 52.
In a further embodiment the peptide fragment under a), the functional
homologue under
b), or the functional analogue under c) activates T cells that recognizes
Arginase 1 expressing
cells. In one embodiment the activation is determined by the ELISPOT assay
described herein. In
a further embodiment the peptide fragment under a), activates T cells that
recognizes Arginase 1
expressing cells as determined by the ELISPOT assay described herein.
In a particular aspect the disclosure concerns a peptide compound of Arginase
1 selected
from:
a) SEQ ID NO 52 or a peptide fragment of SEQ ID NO 52 consisting of a
consecutive sequence
having from 8 to 49 amino acids,
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 52 or
the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 52 or the peptide fragment of a),

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
18
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide;
or a pharmaceutically acceptable salt thereof It is intended that any one of
the peptide
compounds of a), b) or c) can be the subject of individual embodiments.
In a further embodiment the peptide fragment consists of a consecutive
sequence in the
range of from 9 to 49 amino acids, such as from 10 to 40 amino acids, such as
from 20 to 30
amino acids.
In a further embodiment the peptide fragment of SEQ ID NO 52 consisting of a
consecutive sequence having from 8 to 49 amino acids, comprises the SEQ ID NO
51. In a
further embodiment the peptide fragment of SEQ ID NO 52 consisting of a
consecutive sequence
having from 8 to 49 amino acids, comprises the SEQ ID NO 50. In a further
embodiment the
peptide fragment of SEQ ID NO 52 consisting of a consecutive sequence having
from 8 to 49
amino acids, comprises the SEQ ID NO 9.
In a further embodiment the peptide fragment comprises at least one CD4+ and
at least
one CD8+ T cell epitope. In a still further embodiment the peptide fragment
comprises at least
one CD4+ or at least one CD8+ T cell epitope. In a further embodiment the
peptide fragment
comprises at least one CD4+ T cell epitope. In a still further embodiment the
peptide fragment
comprises at least one CD8+ T cell epitope. In a further embodiment the
peptide fragment
comprises all T cell epitopes, in particular all CD4+ and CD8+ epitopes,
located in a hot-spot
region in ARG(161-210). In a still further embodiment the peptide fragment
comprises all T cell
epitopes except one, in particular all CD4+ and CD8+ epitopes, located in a
hot-spot region in
ARG(161-210).
In a further aspect the disclosure relates to a nucleic acid encoding the
peptide compound
of the present invention. The peptide compound is selected from any one of the
above
embodiments. In one embodiment the nucleic acid is selected from the group
consisting of DNA
and RNA.
In a still further aspect the disclosure relates to a vector comprising the
nucleic acid of the
present invention. The nucleic acid is selected from any one of the above
embodiments, and the
peptide compound of the present invention is selected from any one of the
above embodiments.
In one embodiment the vector is selected from a virus vector.
In a further aspect the present disclosure relates to a host cell comprising
the vector of the
present invention. The vector is selected from any one of the above
embodiments, the nucleic

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
19
acid is selected from any one of the above embodiments, and the peptide
compound is selected
from any one of the above embodiments. In one embodiment the host cell is
selected from a
mammalian cell.
In a still further aspect the disclosure relates to a vector comprising a
nucleic acid
encoding an inactive sequence of Arginase 1 selected from:
a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of
from 8 to 321
amino acids,
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 1 or
the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 1 or the peptide fragment of a),
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide,
wherein the nucleic acid expresses the inactive sequence, and wherein the
inactive sequence
comprises at least 1 immunogenic epitope. In one embodiment the epitope is
selected from the
group consisting of SEQ ID NO 2-17, typically the epitope is selected from SEQ
ID NO 9. In
another embodiment the epitope may comprise any sequence of at least 8
consecutive amino
acids comprised within any one of SEQ ID NOs: 50 ¨ 52.
In a further aspect the disclosure relates to a composition comprising the
peptide
compound or the nucleic acid or the vector or the host cell of the present
disclosure, optionally
together with a pharmaceutically acceptable additive, such as carrier or
adjuvants.
In a still further aspect the present disclosure relates to an
immunotherapeutic
composition comprising
a) the peptide compound of the present invention or the nucleic acid of the
present invention or
the vector of the present invention or the host cell of the present invention;
and
b) an adjuvant;
for use as a medicament.
In an embodiment the immunotherapeutic composition is for use in a method for
treatment or prevention of a disease, disorder or condition selected from
cancer. In one
embodiment the cancer is a tumor forming cancer disease. In a further
embodiment the adjuvant
is selected from the group consisting of bacterial DNA based adjuvants,
oil/surfactant based
adjuvants, viral dsRNA based adjuvants, imidazochinilines, and a Montanide ISA
adjuvant.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
In a further aspect the present disclosure relates to a kit-of-parts
comprising;
a) the immunotherapeutic composition of the present invention, and
b) a composition comprising at least one second active ingredient, selected
from an im-
munostimulating compound, such as an interleukin, e.g. IL-2 and or IL-21, an
anti-cancer agent,
5 such as Actimide, Azacitidine, Azathio-prine, Bleomycin, Carboplatin,
Capecitabine, Cisplatin,
Chlorambucil, Cyclophospha-mide, Cytarabine, Daunorubicin, Docetaxel,
Doxifluridine,
Doxorubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine,
Hydroxyurea,
Idarubicin, Irinotec-an, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan,

Mercaptopurine, Metho-trexate, Mitoxantrone, nivolumab, Oxaliplatin,
Paclitaxel,
10 .. pembrolizumab, Pemetrexed, Revlimid, Temozolomide, Teniposide,
Thioguanine, Valrubicin,
Vinblastine, Vincristine, Vindesine and Vinorelbine.
In an embodiment of the kits-of-parts, the provided compositions are to be
administered
simultaneously or sequentially.
In a further aspect the disclsoure relates to a method of treating a clinical
condition
15 characterized by expression of Arginase 1 of SEQ ID NO 1, the method
comprising
administering to an individual suffering from said clinical condition an
effective amount of the
peptide compound of the present invention or the nucleic acid of the present
invention or the
vector of the present invention or the host cell of the present invention.
In a further aspect the disclosure relates to a method of stimulation of
arginase 1 specific
20 .. T-cells, such as CD4 and CD8 T-cells, in a cancer patient, the method
comprising administering
to the cancer patient an effective amount of the peptide compound of, or the
nucleic, or the
vector, or the host cell of the disclosure.
In a still further aspect the present disclosure relates to a method of
suppressing an
immune suppressive function of Arginase 1 expressing cells, in a cancer
patient, the method
comprising administering to the cancer patient an effective amount of the
peptide compound, or
the nucleic acid, or the vector, or the host cell of the disclosure.
In a further aspect the disclosure relates to use of the peptide compound, or
the nucleic
acid, or the vector, or the host cell of the disclosure, for the manufacture
of a medicament, such
as an immunotherapeutic composition or vaccine, for the treatment or
prevention of a cancer
.. characterized by expression of Arginase 1.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
21
In a still further aspect the disclosure relates to the peptide compound, or
the nucleic acid,
or the vector, or the host cell of the disclosure, for use in a method for
treatment or prevention of
a cancer, when administered simultaneously or sequentially with an additional
cancer therapy.
The additional cancer therapy is selected from the group consisting of a
cytokine therapy,
a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor,
chemotherapy,
radiotherapy, immunostimulating substances, gene therapy, anti-bodies and
dendritic cells. In
one embodiment the additional cancer therapy is selected from an immune system
checkpoint
inhibitor, such as a checkpoint blocking antibody (e.g nivolumab,
pembrolizumab), or is selected
from the group consisting of Actimide, Azacitidine, Azathioprine, Bleomycin,
Carboplatin,
Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Dauno-
rubicin,
Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide, Fludarabine,
Fluor-ouracil,
Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide, Leucovorin,
Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone,
Oxaliplatin,
Paclitaxel, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine,
Valrubicin,
Vinblastine, Vincristine, Vindesine and Vinorelbine.
Arginase activity may be measured for instance by using the Arginase Activity
Colorimetric Assay Kit ( BioVision Arginase assay # K755-100). BioVision's
Arginase Activity
Assay kit is simple, sensitive and rapid. In this assay, Arginase reacts with
arginine & undergoes
a series of reactions to form an intermediate that reacts stoichiometrically
with OxiRedTM Probe
to generate the colored product (OD 570 nm). The kit can detect Arginase
activity less than 0.2
U/L in 96-well assay format.
As used herein any amino acid sequence shown may be modified at the C-terminal
amino
acid to be in amide form (-CONH2) or may be in acid form (-COOH), thus any one
of these are
preferred embodiments, and it is intended that any C-terminal amino acid, such
as I, F, R, L, K,
G, M, D, V, S, T, N, Y, P comprises both amide and acid forms unless specified
by -NH2 or -
OH.
The arginase peptide fragments disclosed herein are made by standard peptide
synthesis,
such as solid-phase peptide synthesis (SPPS). SPPS is a standard method for
synthesizing
peptides in the lab. SPPS allows for the synthesis of natural peptides which
are difficult to
express in bacteria, the incorporation of unnatural amino acids,
peptide/protein backbone
modification, and the synthesis of D-proteins, which consist of D-amino acids.
Small porous

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
22
beads are treated with functional units ('linkers') on which peptide chains
can be built. The
peptide will remain covalently attached to the bead until cleaved from it by a
reagent such as
anhydrous hydrogen fluoride or trifluoroacetic acid. The peptide is thus
'immobilized' on the
solid-phase and can be retained during a filtration process while liquid-phase
reagents and by-
products of synthesis are flushed away. The general principle of SPPS is one
of repeated cycles
of deprotection-wash-coupling-wash. The free N-terminal amine of a solid-phase
attached
peptide is coupled to a single N-protected amino acid unit. This unit is then
deprotected,
revealing a new N-terminal amine to which a further amino acid may be
attached. The
superiority of this technique partially lies in the ability to perform wash
cycles after each
reaction, removing excess reagent with all of the growing peptide of interest
remaining
covalently attached to the insoluble resin. There are two majorly used forms
of SPPS ¨ Fmoc and
Boc. Unlike ribosome protein synthesis, solid-phase peptide synthesis proceeds
in a C-terminal
to N-terminal fashion. The N-termini of amino acid monomers is protected by
either of these two
groups and added onto a deprotected amino acid chain. Automated synthesizers
are available for
both techniques, though many research groups continue to perform SPPS
manually. Furthermore,
the skilled person will understand that the processes described above and
hereinafter the
functional groups of intermediate compounds may need to be protected by
protecting group.
When the peptide compounds, nucleic acids, vectors, host cells and
pharmaceutical
compositions herein disclosed are used for the above treatment, a
therapeutically effective
amount of at least one compound is administered to a mammal in need of said
treatment.
As used herein amino acids are identified by the one or three letter code
known to the
person skilled in the art and shown in the table below for convenience:
Amino acids, one and three letter codes
Amino acid Three letter code One letter code
alanine ala A
arginine arg R
asparagine asn N
aspartic acid asp D
asparagine or aspartic acid asx B
cysteine cys C
glutamic acid glu E
glutamine On Q
glutamine or glutamic acid glx Z

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
23
glycine gly
histidine his
isoleucine ile
leucine leu
lysine lys
methionine met
phenylalanine phe
proline pro
serine ser
threonine thr
tryptophan trP
tyrosine tyr
valine val V
The term "treatment" and "treating" as used herein means the management and
care of a
patient for the purpose of combating a condition, such as a disease or a
disorder. The term is
intended to include the full spectrum of treatments for a given condition from
which the patient
is suffering, such as administration of the active compound to alleviate the
symptoms or
complications, to delay the progression of the disease, disorder or condition,
to alleviate or relief
the symptoms and complications, and/or to cure or eliminate the disease,
disorder or condition as
well as to prevent the condition, wherein prevention is to be understood as
the management and
care of a patient for the purpose of combating the disease, condition, or
disorder and includes the
administration of the active compounds to prevent the onset of the symptoms or
complications.
The treatment may either be performed in an acute or in a chronic way. The
patient to be treated
is preferably a mammal; in particular a human being, but it may also include
animals, such as
dogs, cats, cows, monkeys, apes, sheep and pigs.
The term "a therapeutically effective amount" of a peptide compound of the
present
invention or a peptide fragment disclosed herein, as used herein means an
amount sufficient to
cure, alleviate or partially arrest the clinical manifestations of a given
disease and its
complications. An amount adequate to accomplish this is defined as
"therapeutically effective
amount". Effective amounts for each purpose will depend on the severity of the
disease or injury
as well as the weight and general state of the subject. It will be understood
that determining an
appropriate dosage may be achieved using routine experimentation, by
constructing a matrix of
values and testing different points in the matrix, which is all within the
ordinary skills of a
trained physician or veterinary.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
24
In a still further aspect the disclosure relates to a pharmaceutical
composition comprising
the peptide compound, such as peptide fragment, of the present invention and
optionally a
pharmaceutically acceptable additive, such as a carrier or an excipient.
As used herein "pharmaceutically acceptable additive" is intended without
limitation to
.. include carriers, excipients, diluents, adjuvant, colorings, aroma,
preservatives etc. that the
skilled person would consider using when formulating a compound of the present
invention in
order to make a pharmaceutical composition.
The adjuvants, diluents, excipients and/or carriers that may be used in the
composition of
the invention must be pharmaceutically acceptable in the sense of being
compatible with the
peptide compound, peptide fragment, nucleic acid, vector, or host cell and the
other ingredients
of the pharmaceutical composition, and not deleterious to the recipient
thereof It is preferred that
the compositions shall not contain any material that may cause an adverse
reaction, such as an
allergic reaction. The adjuvants, diluents, excipients and carriers that may
be used in the
pharmaceutical composition of the invention are well known to a person within
the art.
Adjuvants are any substance whose admixture into the composition increases or
otherwise modifies the immune response elicited by the composition. Adjuvants,
broadly
defined, are substances which promote immune responses. Adjuvants may also
preferably have
a depot effect, in that they also result in a slow and sustained release of an
active agent from the
administration site. A general discussion of adjuvants is provided in Goding,
Monoclonal
Antibodies: Principles & Practice (2nd edition, 1986) at pages 61-63.
Adjuvants may be selected from the group consisting of: A1K(SO4)2, AlNa(SO4)2,

A1NH4 (SO4), silica, alum, Al(OH)3, Ca3 (PO4)2, kaolin, carbon, aluminum
hydroxide,
muramyl dipeptides, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-DMP), N-
acetyl-
nornuramyl-L-alanyl-D-isoglutamine (CGP 11687, also referred to as nor-MDP), N-

acetylmuramyul-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'2'-dipalmitoyl-sn -
glycero-3-
hydroxphosphoryloxy)-ethylamine (CGP 19835A, also referred to as MTP-PE), RIBI

(MPL+TDM+CWS) in a 2% squalene/Tween-80® emulsion, lipopolysaccharides and
its
various derivatives, including lipid A, Freund's Complete Adjuvant (FCA),
Freund's Incomplete
Adjuvants, Merck Adjuvant 65, polynucleotides (for example, poly IC and poly
AU acids), wax
D from Mycobacterium, tuberculosis, substances found in Corynebacterium
parvum, Bordetella
pertussis, and members of the genus Brucella, Titermax, ISCOMS, Quil A, ALUN
(see US

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
58767 and 5,554,372), Lipid A derivatives, choleratoxin derivatives, HSP
derivatives, LPS
derivatives, synthetic peptide matrixes or GMDP, Interleukin 1, Interleukin 2,
Montanide ISA-51
and QS-21. Various saponin extracts have also been suggested to be useful as
adjuvants in
immunogenic compositions. Granulocyte-macrophage colony stimulating factor (GM-
CSF) may
5 also be used as an adjuvant.
Preferred adjuvants to be used with the invention include oil/surfactant based
adjuvants
such as Montanide adjuvants (available from Seppic, Belgium), preferably
Montanide ISA-51.
Other preferred adjuvants are bacterial DNA based adjuvants, such as adjuvants
including CpG
oligonucleotide sequences. Yet other preferred adjuvants are viral dsRNA based
adjuvants, such
10 as poly I:C. GM-CSF and Imidazochinilines are also examples of preferred
adjuvants.
The adjuvant is most preferably a Montanide ISA adjuvant. The Montanide ISA
adjuvant
is preferably Montanide ISA 51 or Montanide ISA 720.
In Goding, Monoclonal Antibodies: Principles & Practice (2nd edition, 1986) at
pages
61-63 it is also noted that, when an antigen of interest is of low molecular
weight, or is poorly
15 immunogenic, coupling to an immunogenic carrier is recommended. A
peptide compound,
peptide fragment, nucleic acid, vector, or host cell of an immunotherapeutic
composition of the
invention may be coupled to a carrier. A carrier may be present independently
of an adjuvant.
The function of a carrier can be, for example, to increase the molecular
weight of the peptide
compound, peptide fragment, nucleic acid, vector, or host cell in order to
increase activity or
20 immunogenicity, to confer stability, to increase the biological
activity, or to increase serum half-
life. Furthermore, a carrier may aid in presenting the polypeptide or fragment
thereof to T-cells.
Thus, in the immunogenic composition, the polypeptide or fragment thereof may
be associated
with a carrier such as those set out below.
The carrier may be any suitable carrier known to a person skilled in the art,
for example a
25 protein or an antigen presenting cell, such as a dendritic cell (DC).
Carrier proteins include
keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum
albumin, human
serum albumin, thyroglobulin or ovalbumin, immunoglobulins, or hormones, such
as insulin or
palmitic acid. Alternatively, the carrier protein may be tetanus toxoid or
diphtheria toxoid.
Alternatively, the carrier may be a dextran such as sepharose. The carrier
must be
physiologically acceptable to humans and safe.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
26
The immunotherapeutic composition may optionally comprise a pharmaceutically
acceptable excipient. The excipient must be 'acceptable' in the sense of being
compatible with
the other ingredients of the composition and not deleterious to the recipient
thereof. Auxiliary
substances, such as wetting or emulsifying agents, pH buffering substances and
the like, may be
present in the excipient. These excipients and auxiliary substances are
generally pharmaceutical
agents that do not induce an immune response in the individual receiving the
composition, and
which may be administered without undue toxicity. Pharmaceutically acceptable
excipients
include, but are not limited to, liquids such as water, saline,
polyethyleneglycol, hyaluronic acid,
glycerol and ethanol. Pharmaceutically acceptable salts can also be included
therein, for
example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates,
sulfates, and the
like; and the salts of organic acids such as acetates, propionates, malonates,
benzoates, and the
like. A thorough discussion of pharmaceutically acceptable excipients,
vehicles and auxiliary
substances is available in Remington's Pharmaceutical Sciences (Mack Pub. Co.,
N.J. 1991).
The immunotherapeutic composition may be prepared, packaged, or sold in a form
suitable for bolus administration or for continuous administration. Injectable
compositions may
be prepared, packaged, or sold in unit dosage form, such as in ampoules or in
multi-dose
containers containing a preservative. Compositions include, but are not
limited to, suspensions,
solutions, emulsions in oily or aqueous vehicles, pastes, and implantable
sustained-release or
biodegradable formulations. In one embodiment of a composition, the active
ingredient is
provided in dry (for e.g., a powder or granules) form for reconstitution with
a suitable vehicle (e.
g., sterile pyrogen-free water) prior to administration of the reconstituted
composition. The
composition may be prepared, packaged, or sold in the form of a sterile
injectable aqueous or
oily suspension or solution. This suspension or solution may be formulated
according to the
known art, and may comprise, in addition to the active ingredient, additional
ingredients such as
the adjuvants, excipients and auxiliary substances described herein. Such
sterile injectable
formulations may be prepared using a non-toxic parenterally-acceptable diluent
or solvent, such
as water or 1,3-butane diol, for example. Other acceptable diluents and
solvents include, but are
not limited to, Ringer's solution, isotonic sodium chloride solution, and
fixed oils such as
synthetic mono-or di-glycerides.
Other compositions which are useful include those which comprise the active
ingredient
in microcrystalline form, in a liposomal preparation, or as a component of a
biodegradable

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
27
polymer systems. Compositions for sustained release or implantation may
comprise
pharmaceutically acceptable polymeric or hydrophobic materials such as an
emulsion, an ion
exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
Alternatively, the active
ingredients of the composition may be encapsulated, adsorbed to, or associated
with, particulate
carriers. Suitable particulate carriers include those derived from polymethyl
methacrylate
polymers, as well as PLG microparticles derived from poly(lactides) and
poly(lactide-co-
glycolides). See, e.g., Jeffery et al. (1993) Pharm. Res. 10:362-368. Other
particulate systems
and polymers can also be used, for example, polymers such as polylysine,
polyarginine,
polyornithine, spermine, spermidine, as well as conjugates of these molecules.
As mentioned above, the compositions and particularly immunotherapetic
compositions
as herein disclosed may, in addition to the compounds herein disclosed,
further comprise at least
one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier.
In some
embodiments, the pharmaceutical compositions comprise from 1 to 99 weight % of
said at least
one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier
and from 1 to 99
weight % of a compound as herein disclosed. The combined amount of the active
ingredient and
of the pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier
may not constitute
more than 100% by weight of the composition, particularly the pharmaceutical
composition.
In some embodiments, only one compound as herein disclosed is used for the
purposes
discussed above.
In some embodiments, two or more of the compound as herein disclosed are used
in
combination for the purposes discussed above.
The composition, particularly immunotherapetic composition comprising a
compound set
forth herein may be adapted for oral, intravenous, topical, intraperitoneal,
nasal, buccal,
sublingual, or subcutaneous administration, or for administration via the
respiratory tract in the
form of, for example, an aerosol or an air-suspended fine powder. Therefore,
the pharmaceutical
composition may be in the form of, for example, tablets, capsules, powders,
nanoparticles,
crystals, amorphous substances, solutions, transdermal patches or
suppositories.
Further embodiments of the process are described in the experimental section
herein, and
each individual process as well as each starting material constitutes
embodiments that may form
part of embodiments.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
28
The above embodiments should be seen as referring to any one of the aspects
(such as
'method for treatment', 'immunotherapetic composition', 'peptide compound for
use as a
medicament', or 'peptide compound for use in a method') described herein as
well as any one of the
embodiments described herein unless it is specified that an embodiment relates
to a certain aspect or
aspects of the present invention.
All references, including publications, patent applications and patents, cited
herein are
hereby incorporated by reference to the same extent as if each reference was
individually and
specifically indicated to be incorporated by reference and was set forth in
its entirety herein.
All headings and sub-headings are used herein for convenience only and should
not be
construed as limiting the invention in any way.
Any combination of the above-described elements in all possible variations
thereof is
encompassed by the invention unless otherwise indicated herein or otherwise
clearly
contradicted by context.
It is to be understood that different applications of the disclosed products
and methods
may be tailored to the specific needs in the art. It is also to be understood
that the terminology
used herein is for the purpose of describing particular embodiments of the
invention only, and is
not intended to be limiting.
In addition as used in this specification and the appended claims, the
singular forms "a",
"an", and "the" include plural referents unless the content clearly dictates
otherwise. Thus, for
example, reference to "a peptide" includes two or more such peptides.
A "polypeptide" is used herein in its broadest sense to refer to a compound of
two or
more subunit amino acids, amino acid analogs, or other peptidomimetics. The
term
"polypeptide" thus includes short peptide sequences and also longer
polypeptides and proteins.
As used herein, the term "amino acid" refers to either natural and/or
unnatural or synthetic amino
acids, including both D or L optical isomers, amino acid analogs and
peptidomimetics, and any
pharmaceutically acceptable salts thereof.
A "subject" as used herein includes any mammal, preferably a human.
Recitation of ranges of values herein are merely intended to serve as a
shorthand method
of referring individually to each separate value falling within the range,
unless other-wise
indicated herein, and each separate value is incorporated into the
specification as if it were
individually recited herein. Unless otherwise stated, all exact values
provided herein are

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
29
representative of corresponding approximate values (e.g., all exact exemplary
values provided
with respect to a particular factor or measurement can be considered to also
pro-vide a
corresponding approximate measurement, modified by "about," where
appropriate).
All methods described herein can be performed in any suitable order unless
otherwise
indicated herein or otherwise clearly contradicted by context.
The use of any and all examples, or exemplary language (e.g., "such as")
provided
herein, is intended merely to better illuminate the invention and does not
pose a limitation on the
scope of the invention unless otherwise indicated. No language in the
specification should be
construed as indicating any element is essential to the practice of the
invention unless as much is
explicitly stated.
The citation and incorporation of patent documents herein is done for
convenience only
and does not reflect any view of the validity, patentability and/or
enforceability of such patent
documents.
The description herein of any aspect or embodiment of the invention using
terms such as
.. "comprising", "having", "including" or "containing" with reference to an
element or elements is
intended to provide support for a similar aspect or embodiment of the
invention that "consists
of', "consists essentially of', or "substantially comprises" that particular
element or elements,
unless otherwise stated or clearly contradicted by context (e.g., a
composition described herein as
comprising a particular element should be understood as also describing a
composition
consisting of that element, unless otherwise stated or clearly contradicted by
context).
This invention includes all modifications and equivalents of the subject
matter recited in
herein to the maximum extent permitted by applicable law.
ASPECTS
The following are some additional aspects of the present disclosure:
1. A peptide compound of Arginase 1 selected from:
a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of
from 8 to 321
amino acids,
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 1
or the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 1 or the peptide fragment of a),
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide;

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
or a pharmaceutically acceptable salt thereof
2. The peptide compound of aspectl selected from a) a peptide fragment of SEQ
ID NO 1
consisting of a consecutive sequence of from 8 to 321 amino acids, wherein the
C-terminal
5 amino acid also comprises the amide;
or a pharmaceutically acceptable salt thereof
3. The peptide compound of aspect2 wherein the peptide fragment consists of a
consecutive
sequence in the range of from 8 to 300 amino acids, 8 to 250 amino acids, 8 to
200 amino
10 acids, 8 to 150 amino acids, 8 to 120 amino acids, e.g. 10 to 100 amino
acids, 20 to 80 amino
acids, 30 to 60 amino acids, 40 to 50 amino acids.
4. The peptide compound of any one of aspects 2-3 wherein the peptide fragment
of SEQ ID
NO 1 is selected from the group consisting of ARG(1-310), ARG(1-301), ARG(1-
291),
15 ARG(11-322), ARG(21-322), ARG(30-322), ARG(40-322), ARG(11-310), ARG(11-
301),
ARG(11-291), ARG(21-310), ARG(21-301), ARG(21-291), ARG(30-310), ARG(30-301),
ARG(30-291), ARG(40-310), ARG(40-301), and ARG(40-291).
5. The peptide compound of any one of aspects 1-4 wherein the arginase
activity is reduced
20 compared to Arginase 1, preferably reduced to inactivity, as measured by
an arginase activity
assay, such as the Arginase Activity Colorimetric Assay Kit ( BioVision
Arginase assay #
K755-100).
6. The peptide compound of any one of aspects 2-5 wherein the consecutive
sequence
25 comprises one or more sequences selected from any one of SEQ ID NO 2-17,
such as one
sequence selected from SEQ ID NO 9.
7. The peptide compound of any one of aspects 1-6 wherein the peptide
fragment under a), the
functional homologue under b), or the functional analogue under c) activates T
cells that
30 recognizes Arginase 1 expressing cells.
8. The peptide compound of aspect7 wherein the activation is determined by the
ELISPOT
assay described herein.
9. A nucleic acid, such as DNA or RNA, encoding the peptide compound of any
one of the
preceding aspects.
10. A vector, such virus vector, comprising the nucleic acid of aspect9.
11. A host cell, such as mammalian cell, comprising the vector of aspect10.
12. A vector comprising a nucleic acid encoding an inactive sequence of
Arginase 1 selected
from:
a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of
from 8 to 321
amino acids,

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
31
b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to
SEQ ID NO 1
or the peptide fragment of a), and
c) a functional analogue wherein at least one amino acid has been deleted,
inserted and/or
substituted in SEQ ID NO 1 or the peptide fragment of a),
and wherein the C-terminal amino acid of any one of a), b) or c) also
comprises the amide,
wherein the nucleic acid expresses the inactive sequence, and wherein the
inactive sequence
comprises at least 1 immunogenic epitope, such as an epitope selected from the
group
consisting of SEQ ID NO 2-17, typically the epitope is selected from SEQ ID NO
9.
13. A composition comprising the peptide compound of any one of aspects 1-8 or
the nucleic
acid of aspect 9 or vector of aspectl 0 or 12 or host cell of aspectll,
optionally together with
a pharmaceutically acceptable additive, such as carrier or adjuvans.
14. An immunotherapeutic composition comprising
a) the peptide compound of any one of aspects 1-8 or the nucleic acid of
aspect9 or vector
of aspectl 0 or 12 or host cell of aspectll; and
b) an adjuvant;
for use as a medicament.
15. The immunotherapeutic composition of aspect14 for use in a method for
treatment or
prevention of a disease, disorder or condition selected from cancer, such as a
tumor forming
cancer disease.
16. The immunotherapeutic composition of any one of aspects 14-15 wherein the
adjuvant is
selected from the group consisting of bacterial DNA based adjuvants,
oil/surfactant based
adjuvants, viral dsRNA based adjuvants, imidazochinilines, a Montanide ISA
adjuvant.
17. A kit-of-parts comprising;
a) the immunotherapeutic composition of any one of aspects 14-16, and
b) a composition comprising at least one second active ingredient, selected
from an
immunostimulating compound, such as an interleukin, e.g. IL-2 and or IL-21, an
anti-
cancer agent, such as a chemotherapeutic agent, e.g. Actimide, Azacitidine,
Azathioprine,
Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil,
Cyclophosphamide,
Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin,
Etoposide,
Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan,
Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine,
Methotrexate,
Mitoxantrone, nivolumab, Oxaliplatin, Paclitaxel, pembrolizumab, Pemetrexed,
Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine,
Vincristine,
Vindesine and Vinorelbine.
18. The kits-of-parts according to aspect17, where the provided compositions
are to be
administered simultaneously or sequentially.
19. A method of treating a clinical condition characterized by expression of
Arginase 1 of SEQ
ID NO 1, the method comprising administering to an individual suffering from
said clinical

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
32
condition an effective amount of the peptide compound of any one of aspects 1-
8 or the
nucleic acid of aspect9 or vector of aspect10 or 12 or host cell of aspectll.
20. A method of stimulation of arginase 1 specific T-cells, such as CD4 and
CD8 T-cells, in a
cancer patient, the method comprising administering to the cancer patient an
effective
amount of the peptide compound of any one of aspects 1-8 or the nucleic acid
of aspect9 or
vector of aspect10 or 12 or host cell of aspectll.
21. A method of suppressing an immune suppressive function of Arginase 1
expressing cells, in
a cancer patient, the method comprising administering to the cancer patient an
effective
amount of the peptide compound of any one of aspects 1-8 or the nucleic acid
of aspect9 or
vector of aspect10 or 12 or host cell of aspectll.
22. Use of the peptide compound of any one of aspects 1-8 or the nucleic acid
of aspect9 or
vector of aspect10 or 12 or host cell of aspectll for the manufacture of a
medicament, such
as an immunotherapeutic composition or vaccine, for the treatment or
prevention of a cancer
characterized by expression of Arginase 1.
23. A peptide compound of any one of aspects 1-8 or the nucleic acid of
aspect9 or vector of
aspect10 or 12 or host cell of aspectll, for use in a method for treatment or
prevention of a
cancer, when administered simultaneously or sequentially with an additional
cancer therapy,
such as a cytokine therapy, a T-cell therapy, an NK therapy, an immune system
checkpoint
inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene
therapy,
antibodies and dendritic cells.
24. The peptide fragment, nucleic acid, vector or host cell of aspect23
wherein the checkpoint
blocking antibodies are selected from Actimide, Azacitidine, Azathioprine,
Bleomycin,
Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide,
Cytarabine,
Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide,
Fludarabine,
Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide,
Leucovorin,
Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone,
Nivolumab,
Oxaliplatin, Paclitaxel, Pembrolizumab, Pemetrexed, Revlimid, Temozolomide,
Teniposide,
Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine.
The present invention is further illustrated by the following examples that,
however, are
not to be construed as limiting the scope of protection. The features
disclosed in the foregoing
description and in the following examples may, both separately and in any
combination thereof,
be material for realizing the invention in diverse forms thereof.
Examples
Introduction

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
33
The following explains the rationale behind the planning and execution of some
of the
following experiments:
In addition to the new immunogenic epitopes from Arginase 1, and strong immune

responses against the new immunogenic epitopes as well as frequent immune
responses detected
against several peptide fragments of Arginase 1, the present inventor also
will identify if T cells
likewise recognize Arginase. Furthermore, immunogenic epitopes from Arginase
will be
identified. Using the reverse immunology approach, potential MHC class I
binding motifs will
be identified followed by the synthesis of candidate peptides. Subsequently,
the actual binding to
the corresponding MHC alleles will be established. Following the
identification of class I
binding peptide compounds, such peptides will be examined in an ELISPOT assay
for cytokine
release by peptide specific T cells in order to examine whether specific T
cells are present among
peripheral PBMC of cancer patients as well as healthy donors. In the planned
study tetrameric
MHC/-peptide-complexes will be used as additional means of investigating T-
cell reactivity in
peripheral blood by flow cytometry. Finally, tetrameric MHC/peptide complexes
will be used to
isolate specific T cells directly from patient blood or tumor infiltrated
lymph nodes. This will be
used determine the functional capacity of specific T cells.
Additionally, the inventor will search for novel epitopes presented by MHC
class II
molecules, since CD4+ T cells play a critical role in generating and
maintaining antigen-specific
cellular and humoral immune responses. CD4+ T cells are presented with 18-mer
overlapping
synthetic peptides spanning the entire protein sequence in ELISPOT assays.
Peptide-specific
CD4+ T-cell clones are generated by repetitive stimulation with peptide.
Furthermore, using a set
of partially histocompatible EBV-B cell lines and MHC class II-specific
antibodies, the HLA
class II restriction elements will be identified. Finally, tumor infiltrating
lymphocytes (TIL)
cultures will be analyzed for reactivity against CD4 and CD8 peptide epitopes
revealing
.. important information regarding pivotal in vivo targets.
Arginase is a major player in the immune system and is, it could be speculated
that such
T cells are involved in general immune regulation. An additional part of these
studies aim at
analysing the role of Arginase specific T cells may therefore also have a role
in immune
regulation. First, it will be examined if Arginase-specific T cells can effect
immunity by
eliminating arginase-expressing regulatory cells thereby suppressing and/or
delaying local
immune suppression. To examine a possible immune effect of Argiasese T cells,
it will be

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
34
examined if tThe presence of Arginase-specific T cells or the activation of
such T cells by
Arginase- derived peptides may boost additional T- and/or B-cell responses
against other
antigens. Regulatory cells contribute to the strength and duration of a given
immune response.
Thus, any "supportive" effect of Arginase-specific T cells on other immune
cells may well be
mediated in several direct and indirect manners. In this respect, the level of
essential amino acids
will be examined, the frequency of Tregs as well as the frequency of IL-17
producing cells.
Furthermore, the effect of Arginase-specific T cells on the overall production
of different
cytokines including NF-y, IL-6, TNF-a as well as IL-10 and TGF-13, will be
examined. Another
possible effect of Arginase-specific T cells could be mediated through the
metabolites of arginin.
Furthermore, the phenotype of the Arginase-specific T cells by FACS as well as
analysing the
cytokine profile of such T cells will be described. Furthermore, the
expression of co-stimulatory
molecules (FACS), direct or indirect killing of effector cells and APC
(cytotox assays), will be
addressed. The use of leukapheresis samples from cancer patients containing
huge numbers of T
cells will make it possible to perform experiments with a natural subset of
Arginase-specific T
cells directly isolated from donors.
Peptides
The sequences of the peptides used in these experiments are shown in full in
Table A below (see
the section entitled "Sequences"). Peptides are described in Table A by SEQ ID
NO, by name,
or by reference to the start and end positions of each peptide sequence within
the full length
sequence of Arginase 1. Each may be used interchangeably. For example, the
peptide of SEQ ID
NO: 34 may alternatively be referred to by the name Argl-17 or may
alternatively be referred to
as Arg 161-190 (given a start position of 161 and end position of 190). The
intended reference
in each case will be clear from the context. The peptide of SEQ ID NO: 9 may
additionally be
referred to as ArgShort.
20-mer and 22-mer peptides were synthesized by PepScan (Netherlands) and
dissolved in DMSO
at 10mM. 30-mer and 50-mer peptides were synthesized by Schafer-N ApS
(Denmark) and
dissolved in DMSO at 10mM stock. Shorter peptides were synthesized by KJ Ross-
Petersen ApS
(Denmark) and dissolved in sterile water to a stock concentration of 2mM.
Purity of the
synthesized peptides was >80%.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
Example 1
Patients, protocols and methods
Patient material:
5 The present project is based on analyses of blood and tumor lesions from
melanoma, renal cell
carcinoma and ovarian cancer patients. Collection of blood and tumor samples
was conducted at
Herlev Hospital. Blood samples were drawn a minimum of four weeks after
termination of any
kind of anti-cancer therapy. PBMC were isolated using LymphoprepTM (Alere AS,
cat. 1114547)
separation, HLA-typed and frozen in FCS with 10% DMSO (Sigma-Aldrich, cat.
D5879-
10 100ML). The immediate processing of the material was handled at the CCIT
(Centre for Cancer
Immunotherapy) in accordance with local ethical requirements. All patients
only participate upon
written informed consent, and that the ethical committee have approved the
projects.
Enzyme-Linked Immunospot (ELISPOT):
15 Using the "ELISPOT" technique it is possible to screen for T-cell
recognition of a high number
of peptide antigens despite the availability of relatively few T-cells. The
"ELISPOT" technique
takes advantage of the fact that T-cells synthesize cytokines e.g. IFN-y upon
TCR engagement
and subsequent signaling. The method may be used to analyze for secretion of
any cytokine of
choice - in our laboratory we routinely use IFN-y, TNF-a, IL-10, Granzyme-B as
well as perforin
20 - ELISPOT analyses. Standardized quantitation is accomplished by the use
of an ELISPOT
reader (IMMUNOSPOT, CTLanalyzers LLC http://www.immunospot.com/).
Cytotoxicity assay
Conventional 51Cr-release assays for CTL-mediated cytotoxicity were carried
out as described
25 elsewhere [3]. Target cells were T2-cells (ATCC), HLA-A2-1 melanoma cell
lines (FM3 and
FM93)
FACS:
Over the past years, new strong FACS techniques have been developed ¨ in
particular for studies
30 of immune function and specificity. In this respect, peptide/HLA
complexes are readily used for
analyzing the frequency of peptide specific T-cells. Specific T-cells can be
isolated and

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
36
expanded in vitro. For Intracellular stainings, cells were stimulated with
Arginase derived
peptide and for surface markers we employed 4 pl NIR, 10 pl CD4 PerCP, 2 pl
CD8 Pacific
Blue, and 10 pl CD3 MC and for intracellular staining we used 2 pl anti-TNF-a
and anti-IFN-g
antibodies conjugated with either PE-Cy7 or APC. Washing, permeabilization and
staining
procedures followed previously described methods [4]. Flow cytometry analysis
was performed
on a FACSCANTO II (BD Biosciences, San Jose CA, USA)
Results:
16 peptides derived from Arginase 1 were designed. These peptides are referred
to herein as Arg
1 to Arg 16 (SEQ ID NOs: 12 to 17). The sequences of these peptides are shown
in full in Table
A below (see the section entitled "Sequences"). 15 of the peptides (Argl to
Arg15) were
examined in ELISPOT. We scrutinized peripheral blood mononuclear cells (PBMC)
from six
melanoma patients for the presence of specific T-cell responses against Arg-
derived peptide
using the IFN-gamma ELISPOT secretion assay. Strong responses against Arg2
(SEQ ID NO 3),
Arg3 (SEQ ID NO 4), Arg5 (SEQ ID NO 6), Arg 6 (SEQ ID NO 7), Arg7 (SEQ ID NO
8), Arg8
(SEQ ID NO 9), Arg9 (SEQ ID NO 10), Arg 10 (SEQ ID NO 11), Argil (SEQ ID NO
12),
Arg14 (SEQ ID NO 15), Arg15 (SEQ ID NO 16) were detected. Figure 1 exemplifies
Arg-
specific T cell responses. Especially Arg8 (SEQ ID NO 9) was examined in more
detail. Patient
PBMC hosting immune responses towards Arg8. Immune responses (specific
cells/3x10e5 cells)
from 6 cancer patients against Arginase-derived Arg 8 peptide as seen in
figure 2. Very strong
responses were frequently detected among patients PBMC.
We next examined overlapping 20mer overlapping peptides spanning the entire
Arginase
sequence SEQ ID NO 1. Thus there are 30x 20mer peptides and lx 22mer to span
the entire
length of the sequence. The sequences of these peptides are shown in full in
Table A below (see
the section entitled "Sequences") and correspond to SEQ ID NOs: 18 to 48. We
used the above
described peptides to examine immune responses in PMBC from three cancer
patients seen in
figure 3. Frequent immune responses were detected against several peptides
especially the long
peptides referred to as Arg1-23, Argl-18, Argl -27, Argl -17, Argl-19, Argl-
12, Arg1-20, Argl-
4, Argl-1, Arg1-7, Argl-14, Argl -15, Argl -6, Argl-16, Arg1-22, Argl -29. In
addition, we show
responses in PBMC from healthy individuals against these long peptides (figure
4).

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
37
CD8 positive culture can kill target cells loaded with Arg8 on the surface as
examined by
standard chromium release assay as seen in figure 5. Killing of Arginase
positive cancer
(melanoma) cell lines FM3 and FM93 by Arginase specific T cells was also
demonstrated using
standard chromium release assays as seen in figure 6. In this experiment
Arginase specific T
cells were incubated 4 h with either Cr-labeled melanoma cell lines FM3 or
FM93 at different
effector:target ratios. Both cell lines express intracellular Arginase.
Next we examined if CD4 CD4 T-cells can recognize arginase after in vitro
stimulation. Hence,
we chose to analyze PBMC from a patient with a response against Arg8 using
intracellular
cytokine staining. Although this method is less sensitive than ELISPOT, it
allows to elucidate
which immune cells secrete the cytokine identified in ELISPOT. Hence, we
stimulated CD4 T
cells from a cancer patients 5 times with Arg8 peptide. Next, we performed
Intracellular staining
against INF (PE-Cy7A) and TNF-alfa (APC-A) of the T-cell culture after 5
stimulations in vitro
with Arg8 peptide. The culture is either stimulated with or without Arg8
peptide as seen in figure
7. Similar results are seen in Figure 8.
In conclusion: Both CD8 and CD4 T cells can recognize Arginase derived
peptides on the
surface of target cells. The region spanning positions 161 to 190 of Arginasel
appears to be
particularly immunogenic. This region may be referred to herein as a hotspot.
Example 2
Materials and Methods
Additional Peptide stimulation and ELISPOT assay
PBMCs from healthy donors or cancer patients were stimulated with 80pg of
arginase-derived
peptides and 120 U/ml IL-2 (Peprotech, London, UK, cat. 200-02) for a week. 4-
6x105 PBMCs
were then placed in the bottom of ELISPOT plate (nitrocellulose bottomed 96-
well plates by
MultiScreen MAIP N45; Millipore, cat. MSIPN4W50) pre-coated with TN-7 capture
Ab
(Mabtech, cat. 3420-3-1000) and 1-10pg of arginase derived peptides were
added. PBMCs from
each patient and donors were set up in duplicates or triplicates for peptide
and control
stimulations. Cells were incubated in ELISPOT plates in the presence of an
antigen for 14-16

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
38
hours after which they are washed off and secondary biotinylated Ab (Mabtech,
cat. 3420-6-
1000) was added. After 2h incubation unbound secondary antibody was washed off
and
streptavidin conjugated alkaline phosphatase (AP) (Mabtech, cat. 3310-10) was
added for 1 h.
Next, unbound conjugated enzyme is washed off and the assay is developed by
adding
BCIP/NBT substrate (Mabtech, cat. 3650-10). Developed ELISPOT plates were
analysed on
CTL ImmunoSpot S6 Ultimate-V analyzer using Immunospot software v5.1.
Responses were
calculated as the difference between average numbers of spots in wells
stimulated with Arginase-
1 peptides and control wells.
HLA-blocking
For blocking of HLA Class I and II, PBMCs were pre-incubated with 2pg/m1 of
the blocking
antibodies: anti-human HLA-DR, DP, DQ antibody CloneTil39 (Biolegend, cat.
361702) or anti-
human HLA-ABC antibody Clone w6/32 (Dako, Agilent) for 20 min at RT before the
addition of
the peptide.
Establishment of arginase-specific T-cell cultures
Arginase-specific T cell culture were established by stimulation of cancer
patient PBMC with
irradiated ArgShort (SEQ ID NO 9) peptide-loaded autologous DC or PBMCs. The
following
day IL-7 and IL-12 (PeproTech, London, UK, cat. 200-07-10 and 200-12) were
added.
Stimulation of the cultures were carried out every 8 days with ArgShort
peptide loaded irradiated
autologous DC followed by ArgShort peptide-loaded irradiated autologous PBMC.
The day after
peptide stimulation IL-2 (PeproTech, London, UK, cat. 200-12) was added. After
5 stimulations
arginase-specific T cells were enriched using TNF-a cell enrichment kit
(MiltenyiBiotec, cat.
130-091-269).
Generation of dendritic cells
DCs were generated from PBMC by adherence on culture dishes at 37 C for 1-2
hr. in RPMI-
1640. Adherent monocytes were cultured in RPMI-1640 supplemented with 10% FCS
in the
presence of IL-4 (250 U/ml) and GM-CSF (1000 U/ml) (Peprotech, London, UK,
cat. 200-04
and 300-03-100) for 6 days. DCs were matured by addition of IL-13 (1000U/m1),
IL-6

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
39
(1000U/m1) TNF-a (1000U/m1) (Peprotech, London, UK, cat. 200-01B, 200-06 and
300-01A)
and PGE2 (lug/10 (Sigma Aldrich, cat. P6532).
B cell isolation
PBMCs from cancer patients were thawed and rested overnight. B cells were
isolated from
patient PBMCs using Pan B Cell Isolation Kit (Miltenyi Biotec Inc., cat. 130-
101-638) according
to manufacturer's instructions.
Production of in vitro-transcribed mRNA
.. The cDNA encoding Arginase (accession nr. NM 000045 ) was synthesized and
cloned into
either pSP73-SphA64 (kindly provided by Dr. E. Gilboa, Duke University Medical
Center,
Durham, NC) using 5'XhoI/3'PacI restriction sites (Geneart/Life Technologies)
or into the HLA
class II targeting plasmid pGEM-sig-DC.LAMP (kindly by provided by Dr. K.
Thielemans,
Medical School of the Vrije Universiteit Brussel) using 5'BamHI/3'BamHI
restriction sites. Both
plasmids were linearized with SpeI before serving as DNA template for in vitro
transcription.
Electroporation
For mRNA experiments, dendritic cells and B cells were transfected with
Arginase mRNA or
control mRNA encoding GFP or nerve growth factor receptor (NGFR) using
electroporation
parameters as previously described. Briefly, cells were washed twice,
suspended in Opti-MEM
medium (Invitrogen, cat. 11058021) and adjusted to a final cell density of 4-
7x106 cells/ml. The
cell suspension (200-300 ul) was pre-incubated on ice for 5min and 5-10pg of
mRNA was
added. Cell suspension was then transferred into a 4-mm gap electroporation
cuvette and
electroporated. Electroporated cells were further incubated in humidified
atmosphere with 5%
CO2 and used for experimental analysis as specified. Electroporation
efficiency was determined
24 hours later by FACS analysis of the GFP or NGFR transfected cells.
Flow cytometric analysis
Flow cytometry analysis was performed on a FACSCantoTM II (BD Biosciences, San
Jose CA,
USA). Intracellular staining of cell cultures was performed after the cells
were stimulated with
20-mere peptides for 5 hours or 30-mere peptides for 8h (BD GoigiPiugTM cat.
555029, was

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
added after the first hour). The cells were then stained for surface markers,
then washed and
permeabilized by using Fixation/Permeabilization and Permeabilization Buffer
(eBioscience, cat.
00-5123-43), according to manufacturer's instructions. Antibodies used: IFNy-
APC
(cat.341117), TNFa-BV421 (cat.562783), CD4-FITC (cat.347413), CD8- PerCP (cat
.345774)
5 (all from BD Biosciences). Dead cells were stained using FVS510 (564406,
BD Biosciences)
according to manufacturer's instructions.
Results
Spontaneous immune responses against arginase-1
10 We divided the entire arginase-1 protein sequence into overlapping 20-
amino-acid-long peptides,
generating a library o f 31 peptides covering the whole sequence (SEQ ID NOs
18-48)). Each
peptide in the library overlapped with the first 10 amino acids of the
following peptide. Using
this arginase peptide library and the IFNy ELISPOT assay, we next screened
PBMCs from
melanoma patients and healthy donors for spontaneous responses (Fig. 9A and
9B). The PBMCs
15 were stimulated for one week with a pool of 3-4 adjacent 20-mer arginase
library peptides and
low-dose IL-2 (120 U/mL). They were then set up for an IFNy ELISPOT assay to
screen for
responses against each 20-mer peptide separately. The following eight peptides
showed the
highest and most abundant responses in cancer patient PBMCs (peptides labelled
by reference to
start and end position): Arg(31-50), Arg(111-130), Arg(161-180), Arg(171-190),
Arg(181-
20 200), Arg(191-210), Arg(221-240), and Arg(261-280). Among these
overlapping peptides,
Arg(161-180), Arg(171-190), Arg(181-200), and Arg(191-210) spanned a 50-amino-
acid-long
region that was deemed a hot-spot region since nearly all patients harbored a
response against
one or more of these peptides (Fig. 9A). The selected eight peptides were
further used to screen
for spontaneous immune responses against arginase-1 in PBMCs from eight
healthy donors using
25 IFNy ELISPOT. As in the PBMCs from cancer patients, the PBMCs from
healthy donors
showed the highest IFNy responses against the four arginase peptides in the
hot-spot region (Fig.
9B).
Arginase-1 responses in melanoma TILs
30 To investigate the potential presence of arginase-specific T cells among
tumor-infiltrating
lymphocytes in cancer, we screened TILs from eight melanoma patients for
responses against

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
41
arginase-1 derived peptides. To this end, we performed intracellular staining
for IFNy and TNFa
release in response to peptide stimulation. TILs were thawed and rested
overnight without IL-2,
and then stimulated for 5 h with three hot-spot immunogenic region 20-mer
peptides (peptides
labelled by reference to start and end position): Arg(161-180), Arg(171-190),
Arg(181-200). In
one of the T1L cultures, IFNy was released from CD4+ T cells in response to
stimulation with all
three arginase peptides (Fig. 10A), suggesting that the arginase-1 hot-spot
region likely
contained a number of CD4+ T-cell epitopes. The percentage of IFNy-producing
cells was higher
for Arg(171-190) and Arg(181-200) compared to Arg(161-180), and the response
against
Arg(181-200) was almost twice that against Arg(161-180). No 1FNy or TNFa
release was
observed from CD8+ T cells.
Arginase-1 hot-spot region is recognized by CD4+ and CD8+ T cells
Since we most frequently observed responses against arginase-1 peptides from
the hot-
spot region, we next analyzed whether longer peptides covering the same
sequence could be used
.. instead of four 20-mers. A 50-mer peptide covering the entire hot-spot
region elicited lower
responses compared to the 20-mer peptides (data not shown). We then divided
the arginase-1
hot-spot region into two 30-mer peptides that overlapped by 10 amino acids
(peptides labelled by
reference to start and end position): Arg(161-190) and Arg(181-210). These 30-
mer peptides
were used to check for responses in selected PBMCs from cancer patients and
healthy donors,
which had previously shown responses against the 20-mer hot-spot peptides. The
PBMCs were
stimulated for one week with either Arg(161-190) or Arg(181-210) in the
presence of IL-2, and
were then used in IFNy ELISPOT. PBMCs from both cancer patients and healthy
donors showed
high responses against Arg(161-190) (Fig. 10B), comparable to the responses
against 20-mer
peptides. Some PBMCs also showed responses against the 30-mer Arg(181-210)
peptide (Fig.
.. 10C); however, these responses were lower than those against the
overlapping 20-mer peptides
covering the same protein region (Fig. 9B).
To investigate which types of T cells reacted to the peptide epitopes in the
hot-spot
region, we performed intracellular staining for IFNy and TNFa release in PBMCs
from two
healthy donors and one cancer patient, which had shown responses against the
Arg(161-190)
peptide in IFNy ELISPOT. We detected TNFa release from CD4+ T cells after an 8-
hour
incubation with Arg(161-190) (Fig. 11A). We also detected a minor response
from CD8+ T cells

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
42
in some samples (data not shown), suggesting the presence of HLA Class I and
II epitopes in the
arginase-1 hot-spot region. Blocking HLA Class I or Class II expression
partially blocked the
IFNy release in response to Arg(161-190) peptide stimulation, further
demonstrating that
Arg(161-190) contained CD4+ and CD8+ T cell epitopes (Fig. 11B).
We generated an arginase-specific CD4+ T-cell culture by repeated stimulation
of
PBMCs from a melanoma patient with DCs and PBMCs that were loaded with a
minimal
arginase peptide (ArgShort) located in the hot-spot region. The T-cell culture
specific against the
minimal arginase epitope also recognized the 30-mer Arg(161-190) peptide in
IFNy ELISPOT
(Fig. 11C, left) and intracellular staining (Fig. 11C, right). However, the 50-
mer peptide covering
the entire hot-spot region was not recognized. After 8 hours of Arg(161-190)
peptide
stimulation, intracellular staining revealed TNFa release from CD4+ T cells.
T-cell recognition dependent on arginase-1 expression
To assess the ability of arginase-specific CD4+ T cells to recognize and react
against
immune cells producing arginase-1, we transfected autologous dendritic cells
and B cells with
mRNA encoding arginase-1 protein. Autologous DCs were transfected with two
different
constructs encoding arginase-1 mRNA. One of these constructs contained the
arginase-1
sequence fused to the DC-LAMP signal sequence, which targets a protein towards
the lysosomal
compartment and thus directs that protein towards Class II presentation.
Arginase-specific CD4+
T-cell cultures from two different melanoma patients were rested without IL-2
for 24 h, and then
set up for IFNy ELISPOT with electroporated autologous DCs or B cells. We
observed higher
reactivity against DCs and B cells that were transfected with arginase-1 mRNA
compared to
Mock control (Fig. 12A¨D). The responses were even higher against the DCs
transfected with
arginase-DC-LAMP compared to both Mock control and arginase-1 mRNA (Fig. 12E).
After 24
h, we checked the electroporation efficiency of DCs via FACS analysis of
GFP/NGFR-
expressing cells, finding >90% transfection efficiency.
Conclusion
These experiments confirm that the region spanning positions 161 to 190 of
Arginasel is
particularly immunogenic in both cancer patients and healthy donors, giving
rise to both CD4
and CD8 positive T cell responses. The region may contain multiple HLA Class I
and Class II

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
43
restricted. Peptides derived from this region may be particularly effective in
a vaccine against
Arginasel. Such a vaccine would be expected to have treatment benefits in
cancer.
Example 3 ¨ in vivo experiments
Design of peptides for in vivo experiments
In order to design peptides suitable for use in vaccination experiments in
mice, the
sequence of murine Arginasel (SEQ ID NO: 59) was compared to the human
Arginasel
sequence of SEQ ID NO: 1. See alignment shown in Figure 13, which demonstrates
that the
sequences have a high level of similarity. The level of similarity is
particularly high in the
hotspot region of human Arginasel described in the above examples, i.e. the
region
corresponding to positions 161-210 of SEQ ID NO: 1 - this 50 amino acid region
is shown in
bold for both sequences in Figure 13. An alignment of this region and the
corresponding region
in murine Arginase 1 is also shown below:
hArgl: GFSWVTPCISAKDIVYIGLRDVDPGEHYILKTLGIKYFSMTEVDRLGIGK (SEQ ID NO: 52)
mArgl: GFSWVTPCISAKDIVYIGLRDVDPGEHYIIKTLGIKYFSMTEVDKLGIGK (SEQ ID NO: 57)
Only 2 residues of the 50 differ, shown as bold and underlined. A leucine in
the human
sequence is substituted for the highly similar aliphatic amino acid isoleucine
in the mouse, and
an arginine in the human sequence is substituted for the similarly basic
lysine in the mouse. Both
changes are conservative. Accordingly the hotspot region is highly conserved
between humans
and mice.
Given the above similarity, it was determined that relatively few changes
would be
required to create murine analogues of the human peptides tested in the
previous examples. The
peptides used were therefore as follows:
- GFSWVTPCISAKDIVYIGLR (SEQ ID NO: 34)
ARG17 is the sequence of positions 161-180 of human Arginase 1 (SEQ ID NO: 1).
The
corresponding region in murine Arginase 1 is identical. (Also referred to
interchangeably
as ARG1-17 and ARG(161-180) in previous Examples).
- DVDPGEHYILKTLGIKYFSM (SEQ ID NO: 36)
_

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
44
ARG1-19 is the sequence of positions 181-200 human Arginase 1 (SEQ ID NO: 1)..
The
corresponding region in murine Arginasel differs in a single position (the
bold
underlined L is replaced with I in the murine sequence). (Also referred to
interchangeably as ARG(181-200) in previous Examples).
- KDIVYIGL (SEQ ID NO: 53)
mARG1 is the sequence of positions 172-179 of human Arginase 1 (SEQ ID NO: 1).
The
corresponding region in murine Arginase 1 is identical.
- LGIKYFSM (SEQ ID NO: 54)
mARG2 is the sequence of positions 193-200 of human Arginase 1 (SEQ ID NO: 1).
The
corresponding region in murine Arginase 1 is identical.
- KTLGIKYFSMTEVDKLGIGK (SEQ ID NO: 56)
_
mARG20 is the sequence of positions 191-210 of murine Arginase 1 (SEQ ID NO:
59). The
corresponding region in human Arginasel is SEQ ID NO: 37 and differs in a
single position
(the bold underlined K is replaced with R in the human sequence).
Peptide vaccination of C57BL/6 and Balb/c mice
Animals were vaccinated subcutaneously with 100i_tg peptide in DMSO/H20 in a
1:1 emulsion
with incomplete Freund's adjuvant (WA) or montanide. IFA + DMSO/H20 served as
a control
vaccine. Vaccinations were carried out on day() and day7. For subsequent
analysis of the immune
response to specific peptides, mice were sacrificed on day 14 and spleen and
draining lymph
nodes (dLNs) were harvested.
ELISPOT analysis of peptide-specific response
Murine immune cells were subjected to ELISPOT analysis. Single cell suspension
was prepared
from spleen or dLNs by passage through a cell strainer. After lysis of red
blood cells, 0.9x10E6
cells/well were seeded into ELISPOT plates coated with anti-IFNgamma antibody.
Peptide of
interest was added to designated wells and cells were incubated o/n with
peptide. The next day,
cells were removed, plates washed and incubated with biotinylated detection
antibody. Finally,
after addition of Streptavidin-ALP and substrate visible spots appear. Each
spot corresponds to

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
an individual IFNgamma producing cell. Plates were analysed in an Immunospot
analyser and
plotted as spots/0.9x10E6 cells minus background (i.e. minus number of spots
in corresponding
unstimulated (no peptide) wells).
5 Tumor vaccination of male Balb/c mice, age 9-10 weeks, 4 animals per
group
Each animal was inoculated subcutaneously into the left flank with 0.5x10E6
syngeneic
CT26.WT colon cancer cells in 100u1 PBS. On day6 after inoculation when tumors
were
palpable, animals received the first vaccination of peptide or control vaccine
(as described above
for peptide vaccination). On day 13, animals received the second vaccination.
Tumor growth was
10 monitored and tumors were measured 3x per week. Tumor volume was
calculated at V [mm3]=1
x w2 /2 (where 1 is the longest diameter and w perpendicular to 1).
Results
ARG17, ARG1-19 and mARG20 were all found to be immunogenic in both C57BL/6
15 and Balb/c mice. Peptide specific responses were detected in both
C57BL/6 and Balb/c mice
vaccinated with each of ARG17, ARG1-19 and mARG20 (see Figures 14-15). Of the
shorter
peptides, mARG1 was immunogenic in C57BL/6 mice and mARG2 was not immunogenic
in
either strain (data not shown).
In the tumour inoculation experiments, vaccination with each of ARG17, ARG1-19
and
20 mARG20 was found to inhibit tumor growth relative to vaccination with
control peptide. The
effect was most noticeable with ARG1-19 and mARG20, although a treatment
benefit was
apparent for all three tested peptides. This confirms the potential of the
peptides of the invention
(and of vaccination against Arginase 1 in general) as treatments for cancer.
The lower responses to ARG17 may reflect a lack of stability of that peptide,
e.g. due to
25 the presence of a cysteine residue. Replacement of that amino acid by
conservative substitution
could solve this problem without altering other properties of the sequence of
Argl defined by
positions 161-180. However, these results also suggest that the sequence of
Argl defined by
positions 181-200 and particularly 191-210 may be preferred in that they are
easy to
manufacture, stable and appear to generate best responses in mice.

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
46
References
[1] Bronte V, Zanovello P (2005) Regulation of immune responses by L-
arginine metabolism.
Nat Rev Immunol 5:641-54.
[2] Mussai F, De SC, Abu-Dayyeh I, Booth S, Quek L, McEwen-Smith RM, et al.
(2013)
Acute myeloid leukemia creates an arginase-dependent immunosuppressive
microenvironment. Blood 122:749-58.
[3] Andersen MH, Bonfill JE, Neisig A, Arsequell G, Sondergaard I, Valencia G,
et al. (1999)
Phosphorylated Peptides Can Be Transported by TAP Molecules, Presented by
Class I
MHC Molecules, and Recognized by Phosphopeptide-Specific CTL. J Immunol
163:3812-
8.
[4] Ahmad SM, Martinenaite E, Hansen M, Junker N, Borch TH, Met 0, et al. PD-
L1 peptide
co-stimulation increases immunogenicity of a dendritic cell-based cancer
vaccine, in press
ed. 2016.

CA 03039510 2019-04-04
WO 2018/065563 PCT/EP2017/075443
47
SEQUENCES
Full length human Arginase 1 (NP 000036.2) (SEQ ID NO: 1)
MSAKSRTIGI IGAPFSKGQP RGGVEEGPTV LRKAGLLEKL KEQECDVKDY GDLPFADIPN
DSPFQIVKNP RSVGKASEQL AGKVAEVKKN GRISLVLGGD HSLAIGSISG HARVHPDLGV
IWVDAHTDIN TPLTTTSGNL HGQPVSFLLK ELKGKIPDVP GFSWVTPCIS AKDIVYIGLR
DVDPGEHYIL KTLGIKYFSM TEVDRLGIGK VMEETLSYLL GRKKRPIHLS FDVDGLDPSF
TPATGTPVVG GLTYREGLYI TEEIYKTGLL SGLDIMEVNP SLGKTPEEVT RTVNTAVAIT
LACFGLAREG NHKPIDYLNP PK
Region identified as a hotspot for immunogenicity shown bold and underlined
NOTE: the above sequence is isoform2 of human Arginase 1, which has the most
widespread distribution of expression. An
alternative, longer isoforml is expressed predominantly in the liver. The
sequence for this form is provide as NP_001231367.1.
It includes an additional 8 amino acid sequence (VTQNFL IL, SEQ ID NO 6 2 )
inserted between positions corresponding
to positions 43 and 44 of isoform 2 as shown in SEQ ID NO: 1. Any polypeptide
of the invention which spans those positions of
isoform2 may optionally include the insertion of the said additional 8 amino
acids of isoforml.
Full length murine Arginase 1 (NP_031508.1)(SEQ ID NO: 59)
MSSKPKSLEI IGAPFSKGQP RGGVEKGPAA LRKAGLLEKL KETEYDVRDH GDLAFVDVPN
DSSFQIVKNP RSVGKANEEL AGVVAEVQKN GRVSVVLGGD HSLAVGSISG HARVHPDLCV
IWVDAHTDIN TPLTTSSGNL HGQPVSFLLK ELKGKFPDVP GFSWVTPCIS AKDIVYIGLR
DVDPGEHYII KTLGIKYFSM TEVDKLGIGK VMEETFSYLL GRKKRPIHLS FDVDGLDPAF
TPATGTPVLG GLSYREGLYI TEEIYKTGLL SGLDIMEVNP TLGKTAEEVK STVNTAVALT
LACFGTQREG NHKPGTDYLK PPK
Region identified as a hotspot for immunogenicity shown bold and underlined
Full length human Arginase 2 (NP_001163.1)(SEQ ID NO: 60)
MSLRGSLSRL LQTRVHSILK KSVHSVAVIG APFSQGQKRK GVEHGPAAIR EAGLMKRLSS
LGCHLKDFGD LSFTPVPKDD LYNNLIVNPR SVGLANQELA EVVSRAVSDG YSCVTLGGDH
SLAIGTISGH ARHCPDLCVV WVDAHADINT PLTTSSGNLH GQPVSFLLRE LQDKVPQLPG
FSWIKPCISS ASIVYIGLRD VDPPEHFILK NYDIQYFSMR DIDRLGIQKV MERTFDLLIG
KRQRPIHLSF DIDAFDPTLA PATGTPVVGG LTYREGMYIA EEIHNTGLLS ALDLVEVNPQ
LATSEEEAKT TANLAVDVIA SSFGQTREGG HIVYDQLPTP SSPDESENQA RVRI
Region identified as a hotspot for immunogenicity shown bold and underlined

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
48
TABLE A
SEQ Sequence Name Start End
ID NO pos pos
2 QLAGKVAEV Arg1 79 87
3 LVLGGDHSL Arg2 95 103
4 LLKELKGKI Arg3 148 156
ELKGKIPDV Arg4 151 159
6 VMEETLSYL Arg5 211 219
7 HLSFDVDGL Arg6 228 236
8 SLGKTPEEV Arg7 281 289
9 IVYIGLRDV Arg8 174 182
GLLSGLDIM Arg9 268 276
11 DIMEVNPSL Arg10 274 282
12 LLSGLDIMEV Argil 269 278
13 NLHGQPVSFL Arg12 139 148
14 SLVLGGDHSL Arg13 94 103
YLLGRKKRPI Arg14 218 227
16 VLGGDHSLAI Arg15 96 105
17 FLLKELKGKI Arg16 147 156
18 MSAKSRTIGIIGAPFSKGQP Arg1-1 1 20
19 IGAPFSKGQPRGGVEEGPTV Arg1-2 11 30
RGGVEEGPTVLRKAGLLEKL Arg1-3 21 40
21 LRKAGLLEKLKEQECDVKDY Arg1-4 31 50
22 KEQECDVKDYGDLPFADIPN Arg1-5 41 60
23 GDLPFADIPNDSPFQIVKNP Arg1-6 51 70
24 DSPFQIVKNPRSVGKASEQL Arg1-7 61 80
RSVGKASEQLAGKVAEVKKN Arg1-8 71 90
26 AGKVAEVKKNGRISLVLGGD Arg1-9 81 100
27 GRISLVLGGDHSLAIGSISG Arg1-10 91 110
28 HSLAIGSISGHARVHPDLGV Arg1-11 101 120
29 HARVHPDLGVIWVDAHTDIN Arg1-12 111 130
IWVDAHTDINTPLTTTSGNL Arg1-13 121 140
31 TPLTTTSGNLHGQPVSFLLK Arg1-14 131 150
32 HGQPVSFLLKELKGKIPDVP Arg1-15 141 160
33 ELKGKIPDVPGFSWVTPCIS Arg1-16 151 170
34 GFSWVTPCISAKDIVYIGLR Arg1-17 161 180

CA 03039510 2019-04-04
WO 2018/065563
PCT/EP2017/075443
49
35 AKDIVYIGLRDVDPGEHYIL Arg1-18 171 190
36 DVDPGEHYILKTLGIKYFSM Arg1-19 181 200
37 KTLGIKYFSMTEVDRLGIGK Arg1-20 191 210
38 TEVDRLGIGKVMEETLSYLL Arg1-21 201 220
39 VMEETLSYLLGRKKRPIHLS Arg1-22 211 230
40 GRKKRPIHLSFDVDGLDPSF Arg1-23 221 240
41 FDVDGLDPSFTPATGTPVVG Arg1-24 231 250
42 TPATGTPVVGGLTYREGLYI Arg1-25 241 260
43 GLTYREGLYITEEIYKTGLL Arg1-26 251 270
44 TEEIYKTGLLSGLDIMEVNP Arg1-27 261 280
45 SGLDIMEVNPSLGKTPEEVT Arg1-28 271 290
46 SLGKTPEEVTRTVNTAVAIT Arg1-29 281 300
47 RTVNTAVAITLACFGLAREG Arg1-30 291 310
48 LACFGLAREGNHKPIDYLNPPK Arg1-31 301 322
49 GLYITEEIYKTGLLSGLDIM 257 276
50 GFSWVTPCISAKDIVYIGLRDVDPGEHYIL 161 190
51 DVDPGEHYILKTLGIKYFSMTEVDRLGIGK 181 210
52 GFSWVTPCISAKDIVYIGLRDVDPGEHYILKT 161 210
LGIKYFSMTEVDRLGIGK
53 KDIVYIGL mARG1 172 179
54 LGIKYFSM mARG2 193 200
55 DVDPGEHYIIKTLGIKYFSM* 181 200
56 KTLGIKYFSMTEVDKLGIGK* mARG20 191 210
57 GFSWVTPCISAKDIVYIGLRDVDPGEHYIIKT 161 210
LGIKYFSMTEVDKLGIGK*
58 GFSWIKPCISSASIVYIGLRDVDPPEHFILKN
YDIQYFSMRDIDRLGIQK#
* indicates a sequence from murine Arginase 1 which includes at least one
difference relative to the corresponding region of human Arginase1. Residues
which are non-identical with the corresponding human sequence are bold and
underlined. Murine and human Arginase 1 are the same length so start and end
positions are the same.
# indicates a sequence from human Arginase 2. Start and end positions are the
corresponding positions in human Arginase 1.
Alternative human Arginase 1 sequence (SEQ ID NO: 61)
MSAKSRTIGIIGAPFSKGQPRGGVEEGPTVLRAGLLEKLKEQECDVKDYGDLPFADIPN
DSPFQIVKNPRSVGKASEQLAGKVAEVKKNGRISLVLGGDHSLAISIGHARVHPDLGV
IWVDAHTDINTPLTTTSGNLHGQPVSFLLKELKGKIPDVPGFSWVTPCISAKDIVYIGLR
DVDPGEHYILKTLGIKYFSMTEVDRLGIGKVMEETLSYLLGRKRPIHLSFDVDGLDPSF
TPATGTPVVGGLTYREGLYITEEIYKTGLLSGLDIMEVNPSLGKTPEEVTRTVNTAVAIT
LACFGLAREGNHKPIDYLNPPK

Representative Drawing

Sorry, the representative drawing for patent document number 3039510 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2017-10-06
(87) PCT Publication Date 2018-04-12
(85) National Entry 2019-04-04
Correction of Dead Application 2021-12-17
Examination Requested 2022-08-29

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $210.51 was received on 2023-08-30


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-10-07 $100.00
Next Payment if standard fee 2024-10-07 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2019-04-04
Maintenance Fee - Application - New Act 2 2019-10-07 $100.00 2019-04-04
Maintenance Fee - Application - New Act 3 2020-10-06 $100.00 2020-09-08
Maintenance Fee - Application - New Act 4 2021-10-06 $100.00 2022-01-05
Late Fee for failure to pay Application Maintenance Fee 2022-01-05 $150.00 2022-01-05
Request for Examination 2022-10-06 $814.37 2022-08-29
Maintenance Fee - Application - New Act 5 2022-10-06 $203.59 2022-09-01
Maintenance Fee - Application - New Act 6 2023-10-06 $210.51 2023-08-30
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
IO BIOTECH APS
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Maintenance Fee Payment 2022-01-05 1 33
Office Letter 2022-01-05 1 173
Sequence Listing - New Application / Sequence Listing - Amendment / Amendment 2021-11-12 9 328
PCT Correspondence 2021-11-12 9 326
Request for Examination / Amendment 2022-08-29 13 558
Claims 2022-08-29 3 146
Claims 2024-01-05 3 168
Description 2024-01-05 49 3,774
Amendment 2024-01-05 14 612
Abstract 2019-04-04 1 49
Claims 2019-04-04 2 77
Drawings 2019-04-04 15 1,186
Description 2019-04-04 49 2,544
Patent Cooperation Treaty (PCT) 2019-04-04 2 77
Patent Cooperation Treaty (PCT) 2019-04-04 1 41
International Search Report 2019-04-04 6 161
Declaration 2019-04-04 1 12
National Entry Request 2019-04-04 5 176
Cover Page 2019-04-18 1 23
Courtesy Letter 2019-05-21 2 66
Non-Compliance for PCT - Incomplete 2019-09-05 1 52
Examiner Requisition 2023-09-08 3 172

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

No BSL files available.